Skip to main content

Full text of "USPTO Patents Application 10561839"

See other formats


(12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) 



(19) World Intellectual Property Organization 

International Bureau 

(43) International Publication Date 
31 January 2002 (31.01.2002) 




PCT 



(10) International Publication Number 

WO 02/08748 A2 



(51) International Patent Classification 7 : G01N 33/00 
(21) International Application Number: PCT/US0 1/2 1652 



(22) International Filing Date: 

(25) Filing Language: 

(26) Publication Language: 



9 July 2001 (09.07.2001) 
English 
English 



(30) Priority Data: 

60/217,219 
60/217,221 
60/217,666 
60/217,671 
09/804,457 
09/804,458 
09/804,480 
09/804,580 



10 July 2000 
10 July 2000 
10 July 2000 
10 July 2000 
12 March 2001 
12 March 2001 
12 March 2001 
12 March 2001 



(10.07. 
(10.07 
(10.07 
(10.07 
(12.03 
(12.03 
(12.03 
(12.03 



.2000) US 

2000) US 

2000) US 
.2000) US 
.2001) US 
.2001) US 

2001) US 
.2001) US 



P. [US/US]; 10912CorteLuzdelSol, San Diego, CA 92130 
(US). GONZALEZ, Jesus, E., Ill [US/US]; 12442 Caimel 
Cape, San Diego, CA 92130 (US). 

(74) Agent: ARNO, Thomas, R.; Knobbe, Martens, Olson & 
Bear, LLP, 16th Floor, 620 Newport Center Drive, Newport 
Beach, CA 92660 (US). 

(81) Designated States (national): AE, AG, AL, AM, AT, AT 
(utility model), AU, AZ, BA, BB, BG, BR, BY, BZ, CA, 
CH, CN, CO, CR, CU, CZ, CZ (utility model), DE, DE 
(utility model), DK, DK (utility model), DM, DZ, EC, EE, 
EE (utility model), ES, FL FI (utility model), GB, GD, GE, 
GH, GM, HR, HU, ID, IL, IN, IS, JP, KE, KG, KP, KR, KZ, 
LC, LK, LR, LS, LT, LU, LV, MA, MD, MG, MK, MN, 
MW, MX, MZ, NO, NZ, PL, PT, RO, RU, SD, SE, SG, SI, 
SK, SK (utility model), SL, TJ, TM, TR, TT, TZ, UA, UG, 
US, UZ, VN, YU, ZA, ZW. 



(71) Applicant (/or all designated States except US): AURORA (84) Designated States (regional): ARIPO patent (GH, GM, 



BIOSCIENCES CORPORATION [US/US]; HOlOTor- 
reyanaRoad, San Diego, CA 92121 (US). 

(72) Inventors; and 

(75) Inventors/Applicants (for US only): MAHER, Michael, 



KE, LS, MW, MZ, SD, SL, SZ, TZ, UG, ZW), Eurasian 
patent (AM, AZ, BY, KG, KZ, MD, RU, TJ, TM), European 
patent (AT, BE, CH, CY, DE, DK, ES, FI, FR, GB, GR, IE, 
IT, LU, MC, NL, PT, SE, TR), OAPI patent (BF, BJ, CF, 
CG, CI, CM, GA, GN, GW, ML, MR, NE, SN, TD, TG). 

[Continued on next page] 



(54) Title: ION CHANNEL ASSAY METHODS 




(57) Abstract: A method of characterizing the biological activity of a candidate compound may include exposing cells to the can- 
didate compound, and then exposing the cells to a repetitive application of electric fields so as to set the transmembrane potential to 
a level corresponding to a pre-selected voltage dependent state of a target ion channel. 



WO 02/08748 A2 llllllllllllllllllllllllllllllllllllllllllllllllllllllllll 



Published: For two-letter codes and other abbreviations, refer to the "Guid- 

without international search report and to be republished ance Notes on Codes and Abbreviations" appearing at the begin- 
upon receipt of that report ning of each regular issue of the PCT Gazette. 



WO 02/08748 



PCT/US01/21652 



ION CHANNEL ASSAY METHODS 

Cross Reference to Related Applications 
This application claims priority to U.S. Provisional Application Serial Nos. 60/217,671, 
5 entitled INSTRUMENTATION AND METHODS FOR ELECTRICAL STIMULATION, filed on 
July 10, 2000, by Maher et al.; 60/217,666, entitled INSTRUMENTATION AND METHODS 
FOR ELECTRICAL STIMULATION, filed on July 10, 2000, by Mendlein; 60/217,221, entitled 
INSTRUMENTATION AND METHODS FOR ELECTRICAL STIMULATION, filed on July 10, 
2000, by Maher et al.; 60/217,219, entitled INSTRUMENTATION AND METHODS FOR 

10 ELECTRICAL STIMULATION, filed on July 10, 2000, by Maher et al.; and U.S. Application 
Serial Nos. 09/804,457, entitled ION CHANNEL ASSAY METHODS, filed March 12, 2001, by 
Maher et al.; 09/804,480, entitled ION CHANNEL ASSAY METHODS, and filed March 12, 2001, 
by Maher et al.; 09/804,580, entitled HIGH THROUGHPUT METHOD AND SYSTEM FOR 
SCREENING CANDIDATE COMPOUNDS FOR ACTIVITY AGAINST TARGET ION 

15 CHANNELS, filed March 12, 2001, by Maher et al.; and 09/804,458, entitled MULTI-WELL 
PLATE AND ELECTRODE ASSEMBLIES FOR ION CHANNEL ASSAYS, filed 
March 12, 2001. 

Background of the Invention 

Field of the Invention 

20 The present invention relates generally to instrumentation and methods for manipulating 

membrane potentials of living cells via electrical stimulation. 
Description of the Related Art 

It has long been known that the interior of animal and plant cells is electrically negative 
with respect to the exterior. The magnitude of this potential difference is generally between 5 and 

25 90 mV, with most of the potential being developed across the cell membrane. The transmembrane 
potential of a given cell is set by the balance of the activities of ion transporters which create and 
maintain the electrochemical gradient, and the activities of ion channels, passive diffusion and 
other factors, that allow ions to flow through the plasma membrane. 

Ion channels participate in, and regulate, cellular processes as diverse as the generation 

30 and timing of action potentials, energy production, synaptic transmission, secretion of hormones 
and the contraction of muscles, etc. Many drugs exert their specific effects via modulation of ion 
channels. Examples include antiepileptic compounds like phenytoin and lamotrigine, which block 
voltage-dependent sodium channels in the brain, antihypertensive drugs like nifedipine and 
diltiazem, which block voltage-dependent calcium channels in smooth muscle cells, and 

35 stimulators of insulin release like glibenclamide and tolbutamide, which block ATP-regulated 
potassium channels in the pancreas. 



-1- 



WO 02/08748 PCT/US01/21652 

Finding new drugs which have specific modulatory effects on ion channels requires methods 
for measuring and manipulating the membrane potential of cells with the ion channels present in the 
membrane. A number of methods exist today that can be used to measure cell transmembrane 
potentials and to measure the activities of specific ion channels. Probably the best known approach is 
5 the patch clamp, originally developed by Neher, Sakmann, and Steinback. (The Extracellular Patch 
Clamp, A Method For Resolving Currents Through Individual Open Channels In Biological 
Membranes, Pfluegers Arch. 375; 219-278, 1978). Other methods include optical recording of 
voltage-sensitive dyes (Cohen et al., Annual Reviews of Neuroscience 1: 171-82, 1978) and 
extracellular recording of fast events using metal (Thomas et al., Exp. Cell Res. 74: 61-66, 1972) or 

10 field effect transistors (FET) (Fromherz et al., Science 252: 1290-1293, 1991) electrodes. 

The patch clamp technique allows measurement of ion flow through ion channel proteins 
and the analysis of the effect of drugs on ion channels function. In brief, in the standard patch 
clamp technique, a thin glass pipette is heated and pulled until it breaks, forming a very thin (< 1 
jum in diameter) opening at the tip. The pipette is filled with salt solution approximating the 

1 5 intracellular ionic composition of the cell. A metal electrode is inserted into the large end of the 
pipette, and connected to associated electronics. The tip of the patch pipette is pressed against the 
surface of the cell membrane. The pipette tip seals tightly to the cell and isolates a few ion channel 
proteins in a tiny patch of membrane. The activity of these channels can be measured electrically 
(single channel recording) or, alternatively, the patch can be ruptured allowing measurements of 

20 the combined channel activity of the entire cell membrane (whole cell recording). 

During both single channel recording and whole-cell recording, the activity of individual 
channel subtypes can be further resolved by imposing a voltage clamp across the membrane. 
Through the use of a feedback loop, the voltage clamp imposes a user-specified potential 
difference across the membrane, allowing measurement of the voltage, ion, and time dependencies 

25 of various ion channel currents. These methods allow resolution of discrete ion channel subtypes. 

A major limitation of the patch clamp technique as a general method in pharmacological 
screening is its low throughput. Typically, a single, highly trained operator can test fewer than ten 
compounds per day using the patch clamp technique. Furthermore the technique is not easily 
amenable to automation, and produces complex results that require extensive analysis by skilled 

30 electrophysiologists. By comparison, the use of optical detection systems provides for 
significantly greater throughput for screening applications (currently, up to 100,000 compounds 
per day), while at the same time providing for highly sensitive analysis of transmembrane 
potential. Methods for the optical sensing of membrane potential are typically based on 
translocation, redistribution, orientation changes, or shifts in spectra of fluorescent, luminescent, 

35 or absorption dyes in response to the cellular membrane potential (see generally Gonzalez, et al., 
Drug Discovery Today 4:43 1-439, 1999). 



-2- 



WO 02/08748 PCT/US01/21652 

A preferred optical method of analysis has been previously described (Gonzalez and Tsien, 
Chemistry and Biology 4: 269-277, 1997; Gonzalez and Tsien, Biophysical Journal 69: 1272-1280, 
1995; and U.S. Patent No 5,661, 035 issued August 26, 1997). This approach typically comprises 
two reagents that undergo energy transfer to provide a ratiometric fluorescent readout that is 
5 dependent upon the membrane potential. The ratiometric readout provides important advantages 
for drug screening including improved sensitivity, reliability and reduction of many types of 
experimental artifacts. 

Compared to the use of a patch clamp, optical methods of analysis do not inherently 
provide the ability to regulate, or clamp, the transmembrane potential of a cell. Clamping methods 
10 are highly desirable because they provide for significantly enhanced, and more flexible methods of 
ion channel measurement. A need thus exists for reliable and specific methods of regulating the 
membrane potentials of living cells that are compatible with optical methods of analysis and are 
readily amendable to high throughput analysis. 

Summary of the Invention 

15 In one embodiment, a method of assaying ion channel activity comprises exposing at least 

one cell to a plurality of electric field pulses so as to create a controlled change in transmembrane 
potential and so as to activate an ion channel of interest, and detecting ion channel activity by 
detecting one or more changes in transmembrane potential without using a patch clamp. The 
monitoring may comprise detecting fluorescence emission from an area of observation containing 

20 the cells. In some advantageous embodiments, the electric fields are biphasic. 

In another embodiment, the invention comprises a method of characterizing the biological 
activity of a candidate compound. The method includes exposing one or more cells to said 
compound, repetitively exposing said one or more cells to one or more electric fields so as to effect 
a controlled change in transmembrane potential of said one or more cells, and monitoring, without 

25 using a patch clamp, changes in the transmeln one embodiment, a method of assaying ion channel 
activity comprises exposing at least one cell to a plurality of electric field pulses so as to create a 
controlled change in transmembrane potential and so as to activate an ion channel of interest, and 
detecting ion channel activity by detecting one or more changes in transmembrane potential 
without using a patch clamp. 

30 Advantageously, pulsed biphasic electric fields may be used that have a maximum 

amplitude of less than approximately 90 V/cm, are applied at a rate of at least about 1 per second, 
and which have total duration of at least about 1 millisecond. 

In another embodiment, cells are used in an ion channel assay method that express both an 
ion channel of interest and a counter ion channel. 

35 Methods and systems of compound screening are provided. In one embodiment, such a 

method comprises expressing the target ion channel in a population of host cells and placing a 



-3- 



WO 02/08748 PCT/US01/21652 

plurality of the host cells into each of a plurality of sample wells. A candidate drug compound is 
added to at least one of the plurality of sample wells; and the transmembrane potential of the cells 
is modulated with a repetitive application of electric fields so as to set the transmembrane potential 
to a level corresponding to a pre-selected voltage dependent state of the target ion channel. 
5 Apparatus for high throughput screening is also provided. In one specific embodiment, a 

plurality of wells having a high transmittance portion through which cells present in the wells are 
optically observable in an area of observation are each provided with two electrodes. A power 
supply is connected to the electrodes; wherein the power supply and the electrodes are configured 
to apply a series of electric fields to cells within the area of observation, the electric fields having a 

10 spatial variation of less than about 25% of a mean field intensity within the area of observation, the 
electric fields being effective to controllably alter the transmembrane potential of a portion of the 
cells. In addition, an optical detector is configured to detect light emanating from the wells 
through the high transmittance portion, and a data processing unit is provided to interpret the light 
emanating from the wells through the high transmittance portion as ion channel activity resulting 

15 from the transmembrane potential alterations. 

In one embodiment of the invention, an assay plate and electrode assembly comprises at 
least one sample well having electrodes placed therein. The electrodes are positioned with respect 
to the bottom surface of the well to provide an electric field adjacent to the bottom surface that 
varies by less than about 10% from a mean field intensity over at least about 20% of the surface 

20 area of the bottom surface. 

Additional electrode/plate combinations of the invention include a bottom panel for a 
multi-well plate comprising at least one row of high transmittance regions with positions 
corresponding to well locations having a first strip of conductive material extending along the row 
and overlapping a first portion of the well locations, and having a second strip of conductive 

25 material extending along the row and overlapping a second portion of the well locations. 

In another embodiment, an assay apparatus comprises a sample well, a first pair of 
electrodes positioned within the sample well, and at least one additional satellite electrode 
positioned within the sample well. 

Brief Description of the Drawings 

30 FIG. 1 Shows one embodiment of a dipper electrode array. 

FIG. 2 Shows a number of embodiments of multiwell plates comprising surface 
electrodes. 

FIG. 3 Shows a block diagram of one embodiment of the electrical stimulation system. 
FIG. 4. Shows the simulated effects of repetitive external electrical fields on a cell 
35 expressing a voltage dependent sodium channel. The upper panel indicates the applied electrical 



-4- 



WO 02/08748 PCT/US01/21652 

field, the middle panel indicates the simulated sodium current into the cell, and the lower panel 
indicates the simulated average transmembrane potential. 

FIG. 5 Shows a schematic representation of a square wave. 

FIG. 6 Shows examples of various wave kernels. 
5 FIG. 7 Shows calculated electric field profiles for various electrode assemblies in round, 

6.2 mm diameter wells. Dashed circle is a 3 mm diameter view window. In white areas, the 
electric field strength is less than 10% of the average electric field strength in the view window. In 
gray areas, the electric field strength is within 1 0% of the average electric field strength in the view 
window. In black areas, the electric field strength is greater than 1 0% of the average electric field 
1 0 strength in the view window. 

FIG. 8 Shows calculated electric field profiles for various electrode assemblies in round 
and square wells 6.2 mm across. Dashed circle is a 3 mm diameter view window. In white areas, 
the electric field strength is less than 1% of the average electric field strength in the view window. 
In gray areas, the electric field strength is within 1% of the average electric field strength in the 
15 view window. In black areas, the electric field strength is greater than 1% of the average electric 
field strength in the view window. 

FIG. 9 Shows various electrode and insulator designs for improving electric field 
uniformity in round wells. 

FIG. 10 Shows the effect of electrical stimulation protocols at varying pulse amplitudes 
20 over the time course of electrical stimulation in wild-type CHO cells. 

FIG. 11 Shows the relationship between the maximal cellular response and the applied 
pulse amplitude during electrical stimulation for wild-type CHO cells. Data was from FIG. 10 
taken after about 5 seconds. 

FIG. 12 Shows the dose response curve for the effect of TTX in wild-type CHO cells. 
25 Stimulation parameters were 33 V/cm, 50 Hz for 3 seconds with a biphasic square wave kernel (5 
ms per phase). The solid line is a Hill function fit to the data with EC50 = 9 nM and a Hill 
coefficient of 1 .47. 

FIG. 13 Shows the relationship between pulse duration and frequency and the cellular 
response wild-type CHO cells during electrical stimulation. The electric field strength was always 
30 25 V/cm. The stimulus was a three- second burst of biphasic pulses of varying duration and 

Af 

frequency. Solid lines are fits to the form R = 1 H — . 

f fo 

FIG. 14 Shows time traces for CHO cells expressing the NaV2 sodium channel cells 
electrically stimulated at various field strengths. Cells were stimulated in a 96-well plate, with a 
20 Hz, 3 second-long train of biphasic, 5 ms/phase voltage pulses. The stimulation occurred during 
35 the shaded portion of the graph. In this experiment, the cells were stained with 20 \xM CC2-DMPE 



-5- 



WO 02/08748 PCT/US01/21652 

and 63 nM DiSBACg(3). This dye combination has a 2 ms time constant and accurately tracks the 
transmembrane potential. The rise and fall times of the response were fitted to exponential decay 
functions and were found to be x r i se =200 ms and Tf a n =850 ms. 

FIG. 15 Shows the relationship between the electric field strength and the cellular 
5 response measured after 4 seconds (squares) and 10 seconds (circles) of electrical stimulation. The 
line is a Boltzman fit to the data. 

FIG. 16 Shows the effect of pulse duration and stimulation frequency on the cellular 
response of CHO cells expressing the NaV2 sodium channel. 

FIG. 17 Shows the knee time parameter Tq from the fits to the data in FIG. 16 plotted 
1 0 versus the stimulus duration. 

FIG. 18 Shows the temporal response of HEK-293 cells expressing the NaV3 sodium 
channel during electrical stimulation. 

FIG. 19 Shows dose response curves for tetrodotoxin (FIG. 19A) and tetracaine 
(FIG. 19B) for HEK-293 expressing the NaV3 sodium channel. Electrical stimulation conditions 
15 were: E=33 V/cm, 10 ms/phase biphasic stimulation, 15 Hz burst for 1.5 seconds. 

FIG. 20 Shows a dose response curve for tetracaine for HEK-293 expressing the NaV4 ion 
channel. For this experiment, electrical stimulation parameters were E=33 V/cm, 10 ms/phase 
biphasic stimulation, 15 Hz burst for 1.5 seconds. 

FIG. 21. Shows a full-plate view of electrical stimulation of wild-type HEK-293 cells. 
20 Each individual panel represents the time trace of the normalized fluorescence ratio of a single well 
in the 96-well plate. Each well in a vertical column was stimulated simultaneously with the same 
field strength. Field strength increases from left to right. Rows 6-8 contained 10 mM TEA to 
block the voltage-dependent potassium channels. 

FIG. 22. Shows the cellular response as a function of the stimulus field for wild type 
25 HEK. Error bars are standard deviations. Open symbols: no added blockers. Filled symbols: 10 
mM TEA added to block potassium channels. 

FIG. 23 Shows the time response traces for selected concentrations of the sodium channel 
blockers tetrodoxin (TTX) (FIG. 23A) and tetracaine (FIG. 23B) in CHO cells expressing the 
NaV2 sodium channel. 

30 FIG. 24 Shows the dose response curves for TTX and tetracaine inhibition of the NaV2 

sodium channel. 

FIG. 25. Shows a 'Random' TTX spiking experiment. Each small box in this 1 1x8 array 
, contains the ten-second time trace of a well at the corresponding position of a 96-well plate. The 
twelfth column was a control well without cells used for background subtraction and is not shown. 
35 Wells (1,1), (2,2), (3,3), etc. contained a blocking concentration of TTX. 



-6- 



WO 02/08748 PCT/US01/21652 

FIG. 26 Shows an analysis of the 'random 5 TTX spiking data shown in FIG. 25. The data 
points are the ratiometric response in the time window from 1.8-2.4 seconds after the beginning of 
the stimulus burst (i.e. at the peak of the response). The filled circles points were spiked with 1 
jliM TTX; the open circles had no blocker added. 
5 FIG. 27. Shows a full-plate view of electrically-stimulated HL5 cardiac muscle cells. 

Each individual panel represents the time trace of the normalized fluorescence ratio of a single well 
in the 96-well plate. Each well in a vertical column was stimulated simultaneously with the same 
field strength. Field strength increases from left to right. Rows 5 and 6 contained 10 |iM TTX to 
partially block the voltage-dependent sodium channels. Rows 7 and 8 contained 10 raM TEA to 
10 partially block the voltage-dependent potassium channels. 

FIG. 28. Shows the response of HL5 cells as a function of the applied electric field 
strength. Black points are the average of the response of four wells with no added compounds. 
The solid line is a Boltzman fit to the data with E5Q=22 V/cm. The points are the screening 
window: the difference between the response and the unstimulated response normalized to the 
15 standard deviation of the response (see Appendix A3). 

FIG. 29 The typical voltage response for CHO cells expressing a potassium channel and the 
NaV3 sodium channel after a three separate stimulation cycles using surface electrodes. 

FIG. 30 Shows the average ratiometric response of a population of cells grown in a 96 well 
multiwell plate stimulated with monophasic stimuli of varying field strengths via surface electrodes. 
20 The points in this curve are the average peak response of 4 stimulations on the same culture. 

FIG. 31. Shows the cellular response as a function of the stimulus field for wild type 
RBL. Error bars are standard deviations. Open symbols: no added blockers. Filled symbols: 400 
(liM TEA added to block IRK1 channels. 

Detailed Description of the Preferred Embodiment 
25 Generally, the nomenclature used herein and many of the fluorescence, computer, 

detection, chemistry and laboratory procedures described below are those well known and 
commonly employed in the art. Standard techniques are usually used for chemical synthesis, 
fluorescence, optics, molecular biology, computer software and integration. Generally, chemical 
reactions, cell assays and enzymatic reactions are performed according to the manufacturer's 
30 specifications where appropriate. The techniques and procedures are generally performed 
according to conventional methods in the art and various general references, including those listed 
below. 

Lakowicz, J.R. Topics in Fluorescence Spectroscopy, (3 volumes) New York: Plenum 
Press (1991), and Lakowicz, J. R. Emerging applications of fluorescence spectroscopy to cellular 
35 imaging: lifetime imaging, metaUligand probes, multi-photon excitation and light quenching. 
Scanning Microsc Suppl Vol. 10 (1996) pages 213-24, for fluorescence techniques. 



-7- 



WO 02/08748 PCT/US01/21652 

Sambrook et al. Molecular Cloning: A Laboratory Manual, 2 n d ed. (1989) Cold Spring 
Harbor Laboratory Press, Cold Spring Harbor, N.Y., for molecular biology methods. 

Cells: A Laboratory Manual, 1 st edition (1998) Cold Spring Harbor Laboratory Press, 
Cold Spring Harbor, N.Y., for cell biology methods. 
5 Optics Guide 5 Melles Griot® Irvine CA, Optical Waveguide Theory, Snyder & Love 

published by Chapman & Hall for general optical methods. 

Hille, B. Ionic Channels of Excitable membranes, Second Edition (1992) Sinauer 
Associates, Inc., Sunderland, Mass. for general electrophysiological methods and properties of ion 
channels. 

10 Horowitz and Hill, The Art of Electronics, Second Edition (1989) Cambridge University 

Press, Cambridge, U.K. for electronic circuits. 

The following definitions are set forth to illustrate and define the meaning and scope of the 
various terms used to describe the invention herein. 

The term activation refers to the transition from a resting (non-conducting) state of an ion 
15 channel to the activated (conducting) state. 

The term activation threshold refers to the lowest potential above which measurable 
opening of a channel occurs. 

The term anode refers to an electrode when driven to a positive potential relative to earth 
by an external source. 

20 The term area of cellular stimulation means the area defined by two electrodes that 

experiences significant electrical stimulation (typically 5V/cm or higher) in which the cells of 
interest are located. Typically the area of cellular stimulation is larger than, or equal to, the area of 
observation. For standard 96-well based measurements the area of cellular stimulation is typically 
about 16 mm2. 

25 The term area of observation means the portion of the system over which a measurement is 

taken. The area of observation is typically an area of at least 0.5 mm2 for multiwell plate based 
measurements. 

The term bioluminescent protein refers to a protein capable of causing the emission of light 
through the catalysis of a chemical reaction. The term includes proteins that catalyze 
30 bioluminescent or chemiluminescent reactions, such as those causing the oxidation of luciferins. 
The term bioluminescent protein includes not only bioluminescent proteins that occur naturally, 
but also mutants that exhibit altered spectral or physical properties. 

The term biphasic refers to a pulse with two parts, each with an opposite polarity. 



-8- 



WO 02/08748 PCT/US01/21652 

The term Boltzman function refers to the sigmoidal (i.e. step-like) response function 



1 + exp 



x — x 



50 



Ax 

Where: y is the independent variable 

y>0 is an adjustable parameter equal to the limit of the function as x — > oo 

A is an adjustable parameter equal to step size 

X50 is an adjustable parameter related to the midpoint of the step 

Ax is an adjustable parameter describing the width of the step 



The term cathode refers to an electrode when driven to a negative potential relative to earth 
10 by an external source. 

The term depolarize means to cause the transmembrane potential of a cell to become closer 
to zero. In the case of cells that are normally at negative resting potentials, this term means that 
the transmembrane potential changes in a positive direction. 

The term effective concentration (50%) or EC50 refers to the concentration at which a 
15 pharmacological compound has half the effectiveness compared to the maximal effectiveness at 
high concentrations of the compound. 

The term electrically excitable refers to a cell or tissue that responds to a suprathreshold 
electrical stimulus by generating an action potential. Electrically excitable cells contain at least 
one voltage-dependent ion channel type generating an inward current and at least one ion channel 
20 type generating an outward current. 

The term electrical stimulation means initiating a voltage change in cells using an 
extracellular current pulse. 

The term electrode means a controllable conductive interface between an instrument and a 
test system. 

25 The term electropermeablization refers to mild electroporation, in which the hydrated 

pores created through the membrane are only large enough to pass water molecules and small 
single-atom ions. 

The term electroporation refers to a phenomenon in which the application of a large 
electric potential across the membrane of a cell results in dielectric breakdown of the membrane, 
30 and the creation of hydrated pathways through the membrane. 

The term fluorescent component refers to a component capable of absorbing light and then 
re-emitting at least some fraction of that energy as light over time. The term includes discrete 
compounds, molecules, naturally fluorescent proteins and marco-molecular complexes or mixtures 
of fluorescent and non-fluorescent compounds or molecules. The term fluorescent component also 



-9- 



WO 02/08748 PCT/US01/21652 

includes components that exhibit long lived fluorescence decay such as lanthanide ions and 
lanthanide complexes with organic ligand sensitizers, that absorb light and then re-emit the energy 
over milliseconds. 

The term FRET refers to fluorescence resonance energy transfer. For the purposes of this 
5 invention, FRET includes energy transfer processes that occur between two fluorescent 
components, a fluorescent component and a non-fluorescent component, a luminescent component 
and a fluorescent component and a luminescent component with a non-fluorescent component. 

The term gene knockout as used herein, refers to the targeted disruption of a gene in vivo 
with complete loss of function that has been achieved by any transgenic technology familiar to 
10 those in the art. In one embodiment, transgenic animals having gene knockouts are those in which 
the target gene has been rendered nonfunctional by an insertion targeted to the gene to be rendered 
non-functional by homologous recombination. 

The term Hill function refers to the sigmoidal (i.e. step-like) response function 
A 



y( x )= yo 



15 Where: y is the independent variable 

yQ is an adjustable parameter equal to the limit of the function as x — > oo 

A is an adjustable parameter equal to step size 

*0 is an adjustable parameter related to the midpoint of the step 

n is an adjustable parameter describing the steepness of the step 

20 

The term Hill coefficient refers to the parameter n in the Hill function. 
The term hit refers to a test compound that shows desired properties in an assay. 
The term homolog refers to two sequences or parts thereof, that are greater than, or equal 
to 75% identical when optimally aligned using the ALIGN program. Homology or sequence 

25 identity refers to the following. Two amino acid sequences are homologous if there is a partial or 
complete identity between their sequences. For example, 85% homology means that 85% of the 
amino acids are identical when the two sequences are aligned for maximum matching. Gaps (in 
either of the two sequences being matched) are allowed in maximizing matching; gap lengths of 5 
or less are preferred with 2 or less being more preferred. Alternatively and preferably, two protein 

30 sequences (or polypeptide sequences derived from them of at least 30 amino acids in length) are 
homologous, as this term is used herein, if they have an alignment score of more than 5 (in 
standard deviation units) using the program ALIGN with the mutation data matrix and a gap 
penalty of 6 or greater. See Dayhoff, M.O., in Atlas of Protein Sequence and Structure, 1972, 
volume 5, National Biomedical Research Foundation, pp. 101-110, and Supplement 2 to this 

35 volume, pp. 1-10. 



-10- 



WO 02/08748 PCT/US01/21652 

The term hyperpolarize means to cause the transmembrane potential of a cell to move 
farther away from zero. In the case of cells that are normally at negative resting potentials, this 
term means that the transmembrane potential changes in a negative direction. 

The term inactivation means that an ion channel moves into the inactivated state. 
5 The term inactivated refers to a voltage-dependent ion channel in a particular non- 

conducting conformational state. Transitions into and out of the inactivated state are generally 
slow relative to transitions between other conformational states. The inactivated state is usually 
the preferred state at elevated transmembrane potentials. At low transmembrane potentials, the 
inactivated state is unstable and relaxes to the resting state. 

10 The term kernel means a mathematical function intended to be convoluted with one or 

more other time-varying functions. In theory, the kernel can be any function that tends to zero as 
the independent variable tends to ± oo. In practice, the kernel can be any waveform that can 
programmed into an arbitrary wavefunction generator, or that can be generated by a computer- 
controlled digital to analog (D/A) converter. 

15 The term luminescent component refers to a component capable of absorbing energy, such 

as electrical (e.g. Electro-luminescence), chemical (e.g. chemi-luminescence) or acoustic energy 
and then emitting at least some fraction of that energy as light over time. The term component 
includes discrete compounds, molecules, bioluminescent proteins and macro-molecular complexes 
or mixtures of luminescent and non- luminescent compounds or molecules that act to cause the 

20 emission of light. 

The term transmembrane potential modulator refers to components capable of altering the 
resting or stimulated transmembrane potential of a cellular or sub-cellular compartment. The term 
includes discrete compounds, ion channels, receptors, pore forming proteins, or any combination of 
these components. 

25 The term membrane time constant or iy[ means the product of the membrane resistance 

(R-M) and capacitance (Cm)- 

The term monophasic refers to a pulse whose polarity does not change to the opposite 
polarity. 

The term naturally fluorescent protein refers to a protein capable of forming a highly 
30 fluorescent, intrinsic chromophore either through the cyclization and oxidation of internal amino 
acids within the protein or via the enzymatic addition of a fluorescent co-factor. The term includes 
wild-type fluorescent proteins and engineered mutants that exhibit altered spectral or physical 
properties. The term does not include proteins that exhibit weak fluorescence by virtue only of the 
fluorescence contribution of non-modified tyrosine, tryptophan, histidine and phenylalanine groups 
3 5 within the protein. 



-11- 



WO 02/08748 PCT/US01/21652 

The term naturally occurring refers to a component produced by cells in the absence of 
artificial genetic or other modifications of those cells. 

The term Multiwell plate refers to a two dimensional array of addressable wells located on 
a substantially flat surface. Multiwell plates may comprise any number of discrete addressable 
5 wells, and comprise addressable wells of any width or depth. Common examples of multiwell 
plates include 96 well plates, 384 well plates and 3456 well Nanoplates™. 

The term operably linked refers to a juxtaposition wherein the components so described are 
in a relationship permitting them to function in their intended manner. A control sequence 
operably linked to a coding sequence is ligated in such a way that expression of the coding 
10 sequence is achieved under conditions compatible with the control sequences. 

The term polarized cell means a cell with an electric potential difference across its cell 
membrane. 

The term rectification means that the conductance is non-linear, with a preferred direction. 
The term release from inactivation refers to the conversion of an inactivated closed 
15 channel, to a resting closed channel that is now capable of opening. 
The term repetitive means to repeat at least twice. 

The term repolarize means to cause the transmembrane potential of a cell to approach its 
resting potential. 

The term resting or resting state refers to a voltage-dependent ion channel that is closed, 
20 but free from inactivation. 

The term resting potential for a cell means the equilibrium transmembrane potential of a 
cell when not subjected to external influences. 

The term reversal potential for a particular ion refers to the transmembrane potential for 
which the inward and outward fluxes of that ion are equal. 
25 The term substantially parallel means that the distance between the surfaces of two objects 

facing each other varies by less than 10 %, preferably less than 5 %, when measured at every point 
on the relevant surface of each object. 

The term targetable refers to a component that has the ability to be localized to a specific 
location under certain conditions. For example, a protein that can exist at two or more locations 
30 that has the ability to translocate to a defined site under some condition(s) is targetable to that site. 
Common examples include the translocation of protein kinase C to the plasma membrane upon 
cellular activation, and the binding of SH2 domain containing proteins to phosphorylated tyrosine 
residues. The term includes components that are persistently associated with one specific location 
or site, under most conditions. 
35 The term threshold electroporation potential refers to the externally applied field strength 

above which detectable electroporation of a living cell occurs. 



-12- 



WO 02/08748 PCT/US01/21652 

The term test compound refers to a chemical to be tested by one or more screening 
method(s) of the invention as a putative modulator. A test compound can be any chemical, such as 
an inorganic chemical, an organic chemical, a protein, a peptide, a carbohydrate, a lipid, or a 
combination thereof. Usually, various predetermined concentrations of test compounds are used 
5 for screening, such as 0.01 micromolar, 1 micromolar and 10 micromolar. Test compound controls 
can include the measurement of a signal in the absence of the test compound or comparison to a 
compound known to modulate the target. 

The term transformed refers to a cell into which (or into an ancestor of which) has been 
introduced, by means of recombinant nucleic acid techniques, a heterologous nucleic acid 
10 molecule. 

The term transgenic is used to describe an organism that includes exogenous genetic 
material within all of its cells. The term includes any organism whose genome has been altered by 
in vitro manipulation of the early embryo or fertilized egg or by any transgenic technology to 
induce a specific gene knockout. 

15 The term transgene refers any piece of DNA which is inserted by artifice into a cell, and 

becomes part of the genome of the organism (i.e., either stably integrated or as a stable 
extrachromosomal element) which develops from that cell. Such a transgene may include a gene 
which is partly or entirely heterologous (i.e., foreign) to the transgenic organism, or may represent 
a gene homologous to an endogenous gene of the organism. Included within this definition is a 

20 transgene created by the providing of an RNA sequence that is transcribed into DNA and then 
incorporated into the genome. The transgenes of the invention include DNA sequences that encode 
the fluorescent or bioluminescent protein that may be expressed in a transgenic non-human animal. 

The term transistor-transistor logic or TTL refers to an electronic logic system in which a 
voltage around +5V is TRUE and a voltage around 0V is FALSE. 

25 A uniform electric field means that the electric field varies by no more than 15 % from the 

mean intensity within the area of observation at any one time. 

The term voltage sensor includes FRET based voltage sensors, electrochromic 
transmembrane potential dyes, transmembrane potential redistribution dyes, extracellular 
electrodes, field effect transistors, radioactive ions, ion sensitive fluorescent or luminescent dyes, 

30 and ion sensitive fluorescent or luminescent proteins, that are capable of providing an indication of 
the transmembrane potential. 

The following terms are used to describe the sequence relationships between two or more 
polynucleotides: reference sequence, comparison window, sequence identity, percentage identical 
to a sequence, and substantial identity. A reference sequence is a defined sequence used as a basis 

35 for a sequence comparison; a reference sequence may be a subset of a larger sequence, for 
example, as a segment of a full-length cDNA or gene sequence, or may comprise a complete 



-13- 



WO 02/08748 PCT/US01/21652 

cDNA or gene sequence. Generally, a reference sequence is at least 20 nucleotides in length, 
frequently at least 25 nucleotides in length, and often at least 50 nucleotides in length. Since two 
polynucleotides may each (1) comprise a sequence (i.e., a portion of the complete polynucleotide 
sequence) that is similar between the two polynucleotides, and (2) may further comprise a 
5 sequence that is divergent between the two polynucleotides, sequence comparisons between two 
(or more) polynucleotides are typically performed by comparing sequences of the two 
polynucleotides over a comparison window to identify and compare local regions of sequence 
similarity. A comparison window, as used herein, refers to a conceptual segment of at least 20 
contiguous nucleotide positions wherein a polynucleotide sequence may be compared to a 

10 reference sequence of at least 20 contiguous nucleotides and wherein the portion of the 
polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., 
gaps) of 20 percent or less as compared to the reference sequence (which does not comprise 
additions or deletions) for optimal alignment of the two sequences. Optimal alignment of 
sequences for aligning a comparison window may be conducted by the local homology algorithm 

15 of Smith and Waterman (1981) Adv. Appl. Math. 2: 482, by the homology alignment algorithm of 
Needleman and Wunsch (1970) J. Mol. Biol. 48: 443, by the search for similarity method of 
Pearson and Lipman (1988) Proc. Natl. Acad. Sci. (U.S.A.) 85: 2444, by computerized 
implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin 
Genetics Software Package Release 7.0, Genetics Computer Group, 575 Science Dr., Madison, 

20 WI), or by inspection, and the best alignment (i.e., resulting in the highest percentage of homology 
over the comparison window) generated by the various methods is selected. The term sequence 
identity means that two polynucleotide sequences are identical (i.e., on a nucleotide-by-nucleotide 
basis) over the window of comparison. The term percentage identical to a sequence is calculated 
by comparing two optimally aligned sequences over the window of comparison, determining the 

25 number of positions at which the identical nucleic acid base (e.g., A, T, C, G, U, or I) occurs in 
both sequences to yield the number of matched positions, dividing the number of matched 
positions by the total number of positions in the window of comparison (i.e., the window size), and 
multiplying the result by 100 to yield the percentage of sequence identity. The terms substantial 
identity as used herein denotes a characteristic of a polynucleotide sequence, wherein the 

30 polynucleotide comprises a sequence that has at least 30 percent sequence identity, preferably at 
least 50 to 60 percent sequence identity, more usually at least 60 percent sequence identity as 
compared to a reference sequence over a comparison window of at least 20 nucleotide positions, 
frequently over a window of at least 25-50 nucleotides, wherein the percentage of sequence 
identity is calculated by comparing the reference sequence to the polynucleotide sequence which 

35 may include deletions or additions which total 20 percent or less of the reference sequence over the 
window of comparison. 



-14- 



WO 02/08748 PCT/US01/21652 

As applied to polypeptides, the term substantial identity means that two peptide sequences, 
when optimally aligned, such as by the programs GAP or BESTFIT using default gap weights, 
share at least 30 percent sequence identity, preferably at least 40 percent sequence identity, more 
preferably at least 50 percent sequence identity, and most preferably at least 60 percent sequence 
5 identity. Preferably, residue positions which are not identical differ by conservative amino acid 
substitutions. Conservative amino acid substitutions refer to the interchangeability of residues 
having similar side chains. For example, a group of amino acids having aliphatic side chains is 
glycine, alanine, valine, leucine, and isoleucine; a group of amino acids having aliphatic-hydroxyl 
side chains is serine and threonine; a group of amino acids having amide-containing side chains is 

10 asparagine and glutamine; a group of amino acids having aromatic side chains is phenylalanine, 
tyrosine, and tryptophan; a group of amino acids having basic side chains is lysine, arginine, and 
histidine; and a group of amino acids having sulfur-containing side chains is cysteine and 
methionine. Preferred conservative amino acids substitution groups are: valine-leucine-isoleucine, 
phenylalanine-tyrosine, lysine-arginine, alanine- valine, glutamic-aspartic, and asparagine- 

1 5 glutamine. 

Since the list of technical and scientific terms cannot be all encompassing, any undefined 
terms shall be construed to have the same meaning as is commonly understood by one of skill in 
the art to which this invention belongs. Furthermore, the singular forms a, an and the include plural 
referents unless the context clearly dictates otherwise. For example, reference to a restriction 
20 enzyme or a high fidelity enzyme may include mixtures of such enzymes and any other enzymes 
fitting the stated criteria, or reference to the method includes reference to one or more methods for 
obtaining cDNA sequences which will be known to those skilled in the art or will become known 
to them upon reading this specification. 
L Introduction 

25 The present invention recognizes for the first time that the transmembrane potentials of 

intact living cells comprising at least one voltage regulated ion channel, can be precisely 
modulated via the application of repetitive electrical stimulation pulses to the fluid bathing the 
cells. The present invention includes instrumentation and methods that provide for the accurate 
and reliable modulation of the transmembrane potentials of intact living cells without significantly 

3 0 disrupting their native cellular integrity. 

As a non-limiting introduction to the breadth of the invention, the invention includes 
several general and useful aspects, including: 

1) Instrumentation including electrodes, and electrode arrays for reliably generating 
uniform electrical fields in cultures of living cells in aqueous solution. 

35 2) Multiwell plates comprising surface electrodes for high throughput and 

miniaturized stimulation and analysis of ion channel or cellular activities. 



-15- 



WO 02/08748 PCT/US01/21652 

3) Systems for high throughput analysis of ion channel and cellular activities and for 
use in drug discovery, analysis, screening and profiling. 

4) Methods for modulating the transmembrane potential of a living cell via the use of 
repetitive electrical stimulation. 

5 5) Methods for screening the effects of test compounds on the activities of voltage 

regulated, and non-voltage regulated ion channels, transporters and leak currents. Including 
determining state-dependent pharmacological activity of compounds against ion channel and 
transporter proteins. 

6) Methods for profiling and selecting cells or clones based on their response to 
10 electrical stimulation. 

7) Methods for quantitative determination of cellular and ion channel parameters in a 
high-throughput manner, and for quantification of the pharmacological effects of compounds on 
those parameters. 

8) Methods for the introduction of exogenous compounds into the intracellular spaces 

15 of cells. 

9) Methods for modulating the transmembrane potential of intracellular organelles, 
and for screening test compounds against ion channels in these organelles. 

10) Methods for characterizing the physiological effect of the transmembrane potential 
on the function and regulation of physiological and biochemical responses, including gene 

20 expression, enzyme function, protein activity and ligand binding. 

11) Methods for programming or training adaptive neuronal networks or bio- 
computers for specific functional or logical responses. 

12) Methods for providing efficient neuronal interfaces for prosthetic devices 
implanted into an animal, including a human. 

25 These aspects of the invention and others described herein, can be achieved by using the 

methods and instrumentation described herein. To gain a full appreciation of the scope of the 
invention, it will be further recognized that various aspects of the invention can be combined to 
make desirable embodiments of the invention. Such combinations result in particularly useful and 
robust embodiments of the invention. 

30 IL Electrodes and electrode arrays 

In one embodiment, the present invention includes electrodes, and electrode arrays, for 
creating electrical fields across the area of observation. Typically this is achieved via the use of a pair 
of electrically conductive electrodes. An important design feature is that the electrode pairs create 
well-defined electrical fields. Preferred electrode designs include electrode configurations that 

35 maximize the electric field homogeneity experienced by the cells under observation. 



-16- 



WO 02/08748 PCT/US01/21652 

Generating uniform electric fields over the area of observation is important for electrical 
stimulation for several reasons. Firstly, because the cellular response is sensitive to the magnitude 
of the local electric field, non-uniform fields typically cause non-uniform responses in different 
areas, leading to an increased scatter in the results. Secondly, the threshold for 
5 electropermeablization is typically only a factor of 2-5 larger than the transmembrane potentials 
required for electrical stimulation membrane (see Teissie and Rols, 1993, Biophys. J. 65:409-413). 
Thus, if the electric field is too non-uniform, it may not be possible to stimulate all the cells in the 
area of observation without also electropermeablizing some of them. 

Field uniformity over a fixed area can be described in two ways: (1) the standard deviation 
10 of the field magnitude divided by the average field magnitude in the area, and (2) the difference 
between the highest and lowest field magnitudes, normalized to the average field magnitude in the 
area. 

a) Design of Electrodes 

The simplest way to generate a uniform electric field in a conductive medium is to use two 

15 identical, flat electrodes with surfaces that are aligned substantially parallel to each other. 
Generally the closer the electrodes are to each other relative to their width in the transverse 
direction, the greater the field uniformity will be. Typical round multiwell plate wells however 
limit the width of electrodes that can be inserted into the wells, and also introduce two other effects 
which reduce field uniformity. 

20 The roundness of the wells provides a challenge to create a uniform field pointing in one 

direction with two electrodes the width of the conductive saline between the electrodes is 
constantly changing. Additionally the high surface tension of water generates variations in the 
height of the saline across the well when dipper electrodes are inserted. The curved surface, or 
meniscus, can perturb the electric field throughout the volume of the well. The depth of 100 juL of 

25 saline in a 96-well plate is normally about 3.0 mm deep at the center and about 2.9 mm deep at the 
edges of the well. When two stainless steel parallel plate electrodes are inserted, saline is drawn up 
between the electrodes and the walls of the well causing depth variations over the area of 
observation suggesting that the current paths throughout the volume of the saline curve around the 
center, generating electric field non-uniformity. 

30 In one aspect the present invention includes improved electrode designs, and systems for 

electrical stimulation that address these issues to create substantially uniform electrical fields over 
the area of observation. 

In one embodiment, (FIG. 9A) the electrode pair comprises two substantially parallel 
electrodes comprising an electrical insulator that is attached to the pair of electrodes to restrict 

35 current flow to a defined region thereby creating a highly uniform electrical field. 



-17- 



WO 02/08748 PCT/US01/21652 

In another embodiment, (FIG. 9B) the electrode pair additionally comprises satellite 
electrodes to create a more uniform electrical field. 

In another embodiment, (FIG. 9D) the electrode pair is sub-divided into several pieces 
separated by thin insulating dividers. In this case the potential applied to each electrode, expressed 
5 as a fraction of the potential applied to the central most piece can be individually tuned to 
maximize the field uniformity in the area of observation. 

In another aspect, the present invention includes improved electrode designs (FIG. 9C) 
that exhibit improved field uniformity over the area of observation via the elimination or reduction 
of the meniscus effect. 

10 In another aspect multiple electric potential sensors can be fabricated into the surface or 

walls of the wells in a multiwell plate, or attached in arrays to the dipper electrode assembly. 
These sensors can be monitored to manually or automatically adjust the individual electrodes, so as 
to maximize field uniformity. This arrangement will be useful to allow a stimulating electrode 
array to compensate for variations and imperfections in the well shape, volume of saline, variations 

15 in the manufacturing process for the electrodes, damage to the electrode assembly, etc. 

b) Placement of electrodes within the wells 

For dipper electrodes, the ideal situation (in terms of creating a uniform electric field) 
would be to have the bottoms of the electrodes touching the bottom of the well. This way, there 
will be no fringing fields or field non-uniformity associated with vertical current paths. For a 

20 removable structure, however, it is not desirable to require the electrodes to make contact with the 
surface. Small deviations in the plate geometry can cause some electrodes to press into the 
surface, causing damage either to the plate, the cells, or the electrodes. Additionally, in some 
wells, the electrodes may not extend all the way to the surface. For these reasons it may be 
desirable to design a small gap between the bottom of the electrode and the bottom of the well. 

25 Accordingly in one aspect the present invention includes multiwell plates in which the area 

of observation in the middle of the well is raised relative to area around the circumference of the 
well, where the electrodes would be placed. 

The fringing fields will cause non-uniformity over an inter-electrode distance roughly 
equal to the gap between the bottom of the electrode and the bottom of the well. Therefore, this 

30 gap should be kept as small as is practical, preferably in the range of 0.1 to 0.5 mm and the area of 
observation should not typically include any part of the well within this distance from the 
electrodes. 

c) Manufacture of electrodes 

Any electrically conductive material can be used as an electrode. Preferred electrode 
35 materials have many of the following properties, (1) they do not corrode in saline, (2) they do not 
produce or release toxic ions, (3) they are flexible and strong, (4) they are relatively inexpensive to 



-18- 



WO 02/08748 PCT/US01/21652 

fabricate, (5) they are non porous, and (6) they are easily cleaned. Preferred materials include 
noble metals (including gold, platinum, and palladium), refractory metals (including titanium, 
tungsten, molybdenum, and iridium), corrosion-resistant alloys (including stainless steel) and 
carbon or other organic conductors (including graphite and polypyrrole). For many embodiments 
5 stainless steel provides a preferred electrode material. This material is inexpensive, easy to 
machine, and very inert in saline. Stainless steel oxidizes slowly to produce iron oxide when 
passing current in saline, but this does not appear to affect the performance of the system. Iron 
oxide has very low solubility in water and toxic levels of iron do not appear to be released. 
Additionally any iron oxide deposits can easily be removed by soaking the electrodes in 10% nitric 

10 acid in water for two hours, then rinsing thoroughly with distilled water. 

Solid copper and silver electrodes may be used for some applications but are less preferred 
for routine use because they corrode rapidly in saline. Gold plated copper electrodes are relatively 
inert, but appear to lose their gold plating during prolonged electrical stimulation. 

Electrolysis products can be contained or eliminated by coating the surfaces of the electrodes 

15 with protective coatings, such as gelatin, polyacrilimide, or agarose gels. Another potentially useful 
electrode material is an electrochemical half-cell, such as a silver/silver chloride electrode. 

d) Electrode arrays 

Dipper electrodes typically consist of one or more pairs of electrodes that are arranged in an 
array that can be retractably moved into, and out of, one or more wells of a multiwell plate. Dipper 

20 electrodes may be orientated into arrays that match the plate format and density, but can be in arrays 
of any configuration or orientation. For example for a standard 96 well plate, a number of electrode 
configurations are possible including electrode array arrangements to selectively excite one or more 
columns, or rows, simultaneously. 

An example of one embodiment of an electrode array of this type is shown in FIG. 1. In 

25 this example, a 12 by 8 array of electrode pairs is formatted so as to fit into a standard 96-well 
multiwell plate. In this case the electrodes (10) are approximately 4 mm wide, 1 cm long and 0.2 
mm thick, and extend from a conductive comb (50) that is connected through a switch to one side 
of the output stage of a high-power function generator. The electrodes are mounted parallel to 
each other, 4 mm apart, with a non-conductive nylon spacer (20) in between. In this case, the 

30 switch (330) enables one column of the 96 well plate to be selectively stimulated at a time, 
however any temporal, or spatial, combination of stimulation protocols is potentially possible 
given the appropriate configuration of switching, wiring and power function generator. 

The entire array of electrodes is held in correct registration by a rigid non conductive member 
(30) that keeps each electrode pair correctly spaced to accurately match a standard 96 well plate 

35 layout. The non-conductive member (30) provides for the electrodes to move up or down while 
precisely maintaining their registration with the multiwell plate. 



-19- 



WO 02/08748 PCT/US01/21652 

To provide for correct registration of the electrode array with a multiwell plate, the electrode 
assembly can optionally comprise an outer border or flange (40) that can accommodate a standard 96- 
well plate, and enables accurate plate registration. In some embodiments the border (40) can further 
include a registration notch or indentation (80) to provide unambiguous plate registration. 
5 In a preferred embodiment (Also shown in FIG. 1 A) the electrode array further comprises 

means for retractably inserting the electrode array into the wells of the multiwell plate. In one 
embodiment of this configuration, the electrode array further comprises an upper, movable support 
member (90) to which the electrodes (10) are attached. The movable support member (90) is able 
to move up or down relative to the non-conductive member (30) by sliding on four alignment pins 

10 (70). Not shown in these figures is a spring that enables the movable support layer (90) to 
automatically return to the upper position when downward force is no longer applied. A spacer 
(60) provides the ability to lock the movable support layer (90) and electrodes (10) in the fully 
lower orientation. This device allows the electrical stimulator to be used in manual and/or robotic 
screening modes. 

15 III. Multiwell plates for electrical stimulation 

The multiwell plates of the present invention are designed primarily to provide for efficient 
electrical stimulation of cells while at the same time enabling the optical analysis of 
transmembrane potential changes. To accomplish this conductive surface electrodes may be 
orientated in, or on, the walls, bottoms or lids of the multiwell plate. 

20 In general such multiwell plates can have a footprint of any shape or size, such as square, 

rectangular, circular, oblong, triangular, kidney, or other geometric or non-geometric shape. The 
footprint can have a shape that is substantially similar to the footprint of existing multiwell plates, 
such as the standard 96-well microtiter plate, whose footprint is approximately 85.5 mm in width 
by 127.75 mm in length, or other sizes that represent a current or future industry standard (see T. 

25 Astle, Standards in Robotics and Instrumentation, J. of Biomolecular Screening, Vol. 1 pages 163- 
168,1996). Multiwell plates of the present invention having this footprint can be compatible with 
robotics and instrumentation, such as multiwell plate translocators and readers as they are known 
in the art. 

Typically, wells will be arranged in two-dimensional linear arrays on the multiwell plate. 
30 However, the wells can be provided in any type of array, such as geometric or non-geometric 
arrays. The multiwell plate can comprise any number of wells. Larger numbers of wells or 
increased well density can also be easily accommodated using the methods of the claimed 
invention. Commonly used numbers of wells include 6, 12, 96, 384, 1536, 3456, and 9600. 

Well volumes typically can vary depending on well depth and cross sectional area. 
35 Preferably, the well volume is between about 0. 1 microliters and 500 microliters. 



-20- 



WO 02/08748 PCT/US01/21652 

Wells can be made in any cross sectional shape (in plan view) including, square, round, 
hexagonal, other geometric or non-geometric shapes, and combinations (intra-well and inter-well) 
thereof. Preferred are square or round wells, with flat bottoms. 

The walls can be chamfered (e.g. having a draft angle). Preferably, the angle is between 
5 about 1 and 10 degrees, more preferably between about 2 and 8 degrees, and most preferable 
between about 3 and 5 degrees. 

The wells can be placed in a configuration so that the well center-to well-center distance 
can be between about 0.5 millimeters and about 100 millimeters. The wells can be placed in any 
configuration, such as a linear-linear array, or geometric patterns, such as hexagonal patterns. The 
10 well-to- well distance can be about 9 mm for a 96 well plate. Smaller well-center to well-center 
distances are preferred for smaller volumes. 

The wells can have a depth between about 0.5 and 100 millimeters. Preferably, the well 
depth is between about 1 millimeter and 100 millimeters, more preferably between about 2 
millimeters and 50 millimeters, and most preferably between about 3 millimeters and 20 
15 millimeters. 

The wells can have a diameter (when the wells are circular) or maximal diagonal distance 
(when the wells are not circular) between about 0.2 and 100 millimeters. Preferably, the well 
diameter is between about 0.5 and 100 millimeters, more preferably between about 1 and 50 
millimeters, and most preferably, between about 2 and 20 millimeters. 

20 The multiwell plate, will generally be composed of electrically non-conductive material 

and can comprise an optically opaque material that can interfere with the transmission of radiation, 
such as light, through the wall of a well or bottom of a well. Such optically opaque materials can 
reduce the background associated with optical detection methods. Optically opaque materials can 
be any known in the art or later developed, such as dyes, pigments or carbon black. The optically 

25 opaque material can prevent radiation from passing from one well to another, to prevent cross-talk 
between wells, so that the sensitivity and accuracy of the assay is increased. The optically opaque 
material can also be reflective, such as those known in the art, such as thin metal layers, mirror 
coatings, or mirror polish. Optically opaque materials can be coated onto any surface of the 
multiwell plate, or be an integral part of the plate or bottom as they are manufactured. Optically 

30 opaque material can prevent the transmittance of between about 100% to about 50% of incident 
light, preferably between about 80% and greater than 95%, more preferably greater than 99%. 

Since most measurements will not typically require light to pass through the wall of the 
well, materials such as polymers can include pigments to darken well walls or absorb light. Such 
application of pigments will help reduce background fluorescence. Pigments can be introduced by 

35 any means known in the art, such as coating or mixing during the manufacture of the material or 
multiwell plate. Pigment selection can be based on a mixture of pigments to dampen all 



-21- 



WO 02/08748 PCT/US01/21652 

background inherent to the polymer, or a single pigment or ensemble of pigments selected to filter 
or absorb light at desired wavelengths. Pigments can include carbon black. 

Surface electrodes can be embedded or otherwise attached to the wall in a variety of 
formats and arrangements, for example as several narrow vertical electrode stripes. By 
5 appropriately tuning the relative potentials of each stripe, uniform electric fields can be generated 
in the area of observation. Further, using a circular insert, or by embedding vertical stripe 
electrodes all around the well, uniform electrical fields can be generated in any direction across the 
well. It would also be possible to create a uniform field in one direction, followed by a uniform 
field in another direction. This could be useful for cell types whose electrical characteristics are 

10 anisotropic, such as neural or muscle cells, or for cell types with large aspect ratios. 

Each well also comprises a bottom having a high transmittance portion and having less 
fluorescence than a polystyrene-bottom of at least about 90 percent of said bottom's thickness. 
This property can be determined by comparing the fluorescence of an appropriate control bottom 
material with the fluorescence of a test material. These procedures can be performed using well 

15 known methods. Preferably, the bottom is a plate or film as these terms are known in the art. The 
thickness of the bottom can vary depending on the overall properties required of the plate bottom 
that may be dictated by a particular application. Such properties include the amount of intrinsic 
fluorescence, rigidity, breaking strength, and manufacturing requirements relating to the material 
used in the plate. Well bottom layers typically have a thickness between about 10 micrometers 

20 and about 1000 micrometers. Preferably, the well bottom has a thickness between about 10 
micrometers and 450 micrometers, more preferably between about 15 micrometers and 300 
micrometers, and most preferably between about 20 micrometers and 100 micrometers. 

The bottom of a well can have a high transmittance portion, typically meaning that either 
all or a portion of the bottom of a well can transmit light. The bottom can have an optically opaque 

25 portion and a high transmittance portion of any shape, such as circular, square, rectangular, kidney 
shaped, polygonal, or other geometric or non-geometric shape or combinations thereof. 

Preferably, the bottom of the multiwell plate can be substantially flat, e.g. having a surface 
texture between about 0.001 mm and 2 mm, preferably between about 0.01 mm and 0.1 mm (see, 
Surface Roughness, Waviness, and Lay, Am. Soc. of Mech. Eng. #ANSI ASME B46. 1-2985 

30 (1986)). If the bottom is not substantially flat, then the optical quality of the bottom and wells can 
decrease because of altered optical and physical properties of one or both. 

For surface electrode embodiments, the bottom will preferably comprise strips of 
electrically conductive material or coatings that overlap the edge of the wells of the multiwell plate 
and are in electrical contact with the contents of the wells. The electrically conductive strips will 

35 typically terminate at electrical connectors to enable facile attachment to the output stage of a high- 
power function generator as described previously. The electrically conductive strips should have 



-22- 



WO 02/08748 PCT/US01/21652 

low enough resistance so that they can carry the stimulating currents without excessive loss in voltage 
over their length. The resistance from the connector end to the farthest well end should be less than 
10 and more preferably less than 1 £2, and more preferably still less than 0.1 £1 The cross- 
sectional area of the electrically conductive strips should be large enough to accomplish the resistance 
5 requirement For commonly employed electrical conductors, this cross sectional area should be at 
least 10 _ 4 mm^ 5 and more preferable at least 10"3 rnrn^, 

In practice, any conductive materials could be used as long as they are capped with a 
conductive material that is inert in saline. Such materials include the noble metals (including gold, 
platinum, and palladium) and the refractory metals (including chromium, molybdenum, iridium, 

10 tungsten, tantalum, and titanium) as well as alloys thereof. Preferred materials for the conductive 
material for surface electrodes include combinations of chromium, copper, gold, and indium-tin-oxide 
that can be readily embedded or electroplated into or on the transparent bottom layer. Electrolysis 
products can be contained or eliminated by coating the surfaces of the electrodes with protective 
coatings, such as gelatin, polyacrilimide, or agarose gels. 

15 Another potentially useful electrode material is an electrochemical half-cell, such as a 

silver/silver chloride electrode. 

The electrically conductive material coatings or surface modifications can be introduced 
into the bottom using any suitable method known in the art, including vacuum deposition, 
electroplating, printing, spraying, radiant energy, ionization techniques or dipping. Surface 

20 modifications can also be introduced by appropriately derivatizing a polymer or other material, 
such as glass or quartz, before, during, or after the multiwell plate is manufactured and by 
including an appropriate derivatized polymer or other material in the bottom layer. The derivatized 
polymer or other material can then be reacted with a chemical moiety that is used in an application 
of the plate. Prior to reaction with a chemical moiety, such polymer or other material can then 

25 provide either covalent or non-covalent attachment sites on the polymer or other material. Such 
sites in or on the polymer or other material surface can be used to attach conductive layers to the 
plates. Examples of derivatized polymers or other materials include those described by U.S. Patent 
5,583,21 1 (Coassin et al.) and others known in the art or later developed. 
Materials and manufacturing 

30 The materials for manufacturing the multiwell plate will typically be polymeric, since 

these materials lend themselves to mass manufacturing techniques. However, other materials can 
be used to make the bottom of the multiwell plate, such as glass or quartz. The bottom can be 
made of the same or different materials and the bottom can comprise polystyrene, or another 
material. Preferably, polymers are selected that have low fluorescence and or high transmittance. 

35 Polymeric materials can particularly facilitate plate manufacture by molding methods known in the 
art and developed in the future, such as insert or injection molding. 



-23- 



WO 02/08748 PCT/US01/21652 

The multiwell plate of the present invention can be made of one or more pieces. For 
example, the plate and bottom can be made as one discrete piece. Alternatively, the plate can be 
one discrete piece, and the bottom can be a second discrete piece, which are combined to form a 
multiwell plate. In this instance, the plate and bottom can be attached to each other by sealing 
5 means, such as adhesives, sonic welding, heat welding, melting, insert injection molding or other 
means known in the art or later developed. The plate and bottom can be made of the same or 
different material. For example, the plate can be made of a polymer, and the bottom made of 
polystyrene, cycloolefin, Aclar, glass, or quartz. 

Miniaturized surface electrode designs are feasible in standard plate formats (96, 384, 1536) 

10 as well as 3456 and higher plate densities. The throughput of such systems is potentially extremely 
high. For example, assuming 3456 wells per plate screened at 30 plates per hour corresponds to an 
overall throughput of approximately 800,000 wells per eight-hour day, which is approximately 8 
times faster than is presently available, assuming equal plate read times. 

An example of one embodiment of multiwell plate with surface electrodes is shown in 

15 FIG. 2A. In this example, pairs of conductive strips (200) are attached in parallel to an optically 
transparent bottom layer (210) such as glass, or plastic such as COC (see U.S. Patent Number 
5,910,287, issued June 8, 1999) in a 96-well plate format. In this example, the strips of conductive 
material (200) are approximately 2 mm wide, 10 urn thick, and separated by distance of 
approximately 4 mm to enable optical analysis of the cells located in the wells (220), between the 

20 electrodes through the optically transparent bottom layer (210). In other embodiments the strips of 
conductive material can comprise stainless steel wires (from about 0.001 to about 0.010 diameter). 
The optically transparent bottom layer (210) is attached to a 96-well multiwell plate array (230) and 
replaces the normal plate bottom. The strips of electrically conductive material (200) overlap the 
edge of the wells (220) of the 96-well multiwell plate and are in electrical contact with the contents of 

25 the wells. The electrically conductive strips (200) terminate at electrical contacts (240) to enable 
facile attachment to the output stage of a high-power function generator as described previously. In 
this example, there are two electrode contacts per eight-well column in the first well of the column. 
This permits the use of standard 96-well plate layouts, for simpler handling during cell culturing. No 
cells or saline are inserted into these wells. This design permits the simultaneous stimulation of seven 

30 wells in a single column. During the assay, the operator or a robot will temporarily attach wires to the 
contacts, for example with push-pin test electrodes. 

Another embodiment of a multwell plate with surface electrodes is shown in FIG. 2B. In 
this embodiment, the transparent bottom layer (210) extends beyond the edge of the multiwell plate 
(230). In this configuration, all wells remain available for use with cells and compounds. Further, 

35 attachment of external wiring to the contacts (240) is simplified. Push-pin contacts, circuit-board 
edge connectors, or zero- insertion force sockets can be used to make contact with the electrodes. The 



-24- 



WO 02/08748 PCT/US01/21652 

extended bottom layer (210) may make the plates less convenient to manipulate during routine use. 
This can be remedied by bringing the electrode traces (200) to the reverse side of the bottom layer 
(210) during the manufacturing process. This can be accomplished by several methods. For 
example, using two-sided processing of the plates to create contact areas, through-holes can be made 
5 and electroplated, or conducting traces can be wrapped around the edge of the bottom layer. As 
another example, the bottom layer can be made of a flexible insulating material. Then, after making 
the structure as shown in FIG. 2B 5 the part of the bottom layer which protrudes from the edge of the 
plate can be folded and attached to the underside of the plate. 

Another embodiment of a multwell plate with surface electrodes is shown in FIG. 2C. In 

10 this embodiment, the electrodes (200) are attached to the contact pads (240) with narrow via wires 
(205). This permits the use of standard 96-well plate layouts, for simpler handling during cell 
culturing. In this embodiment, all of the electrodes of one polarity are shorted together. Selection of 
a single column is accomplished by supplying the current pulse to only one electrode of the other 
polarity. In this embodiment, no cells, saline, or compounds are placed into the final column where 

1 5 the contact pads are. During the assay, the operator or a robot will temporarily attach wires to the 
contacts, for example with push-pin test electrodes. 

Another embodiment of a multwell plate with surface electrodes is shown in FIG. 2D. In 
this embodiment, the electrodes (200) are aligned parallel to the longer dimension of the 96-well 
plate. This design is essentially similar to the design shown in FIG. 2A, with the exception that 

20 eleven wells in a row will be simultaneously stimulated. 

Preferred materials for the conductive material for surface electrodes include combinations of 
chromium, copper, gold, and indium-tin-oxide that can be readily embedded, attached, or 
electroplated into or on the transparent bottom layer. In practice, any conductive materials could be 
used as long as they are capped with a conductive material that is inert in saline. Such inert materials 

25 include the noble metals (including gold, platinum, and palladium), the refractory metals (including 
chromium, molybdenum, iridium, tungsten, tantalum, and titanium), corrosion-resistant alloys 
(including stainless steel), and carbon or other organic conductors (including graphite and 
polypyrrole) as well as combinations or alloys of these materials. 
IV. Systems for Electrical Stimulation and Spectroscopic Measurement 

30 The present invention includes systems for automated electrical stimulation and 

spectroscopic measurement, comprising: at least one electrode assembly, a means for electrical 
stimulation, an optical detector, and computer control means to coordinate the generation of 
electrical stimuli, collection of data and movement of multiwell plates. The system can further 
comprise means for fluid addition. In one aspect these systems are designed for modulating, 

35 characterizing and assaying the activity of ion channels, transporters, leak currents present in or on 
the surfaces of living cells, and for rapidly screening for the effects of test compounds on the 



-25- 



WO 02/08748 PCT/US01/21652 

effects of channel or cellular activities. The present invention is also directed to chemical entities 
and information (e.g., modulators or chemical or biological activities of chemicals) generated or 
discovered by operation of workstations of the present invention. 

FIG. 3 shows a block diagram of the major electrical and optical components for one 
5 embodiment of a system for automated electrical stimulation and spectroscopic measurement. In 
this example a 96-well multiwell plate dipper electrode array (FIG. 1) was used for electrical 
stimulation. In addition to the stimulator electrode array, the system has several additional 
electrical, optical and mechanical components, as described in detail in commonly owned U.S. 
Patent Application No. 09/1 18,728, filed July 24, 1998. 

10 In this embodiment, a National Instruments (Austin, TX) PC-DIO 24 digital input/output 

card on board the computer (300) is used to set the proper channel on a l-to-12 switch (330) 
(National Instruments ER-16). The computer controlling the fluorescent plate reader (300) also 
sends out a TTL signal to trigger the function generators (310) when the stimulus is programmed to 
begin. Stimulus signals are generated by two arbitrary waveform generators (310). The function 

15 generators are Tektronix (Beaverton, OR) model number AFG310. The first triggers a series of 
TTL pulses to the second which is programmed with the individual stimulus waveform. More 
complex waveform trains can be generated by connecting multiple waveform generators in series 
and/or in parallel. These waveform generators would be triggered by the computer-generated TTL 
pulse or by each other. Alternatively, an A/D converter or a sound card on board the computer 

20 could be used to generate a train of stimuli. In this case, commercially-available or custom 
software could be used to program the waveform train, or to change the waveform during the train. 

The train of stimuli is sent through a high-power amplifier (320), through the switch 
(330), and into the stimulator head (370). In this case the amplifier was built using the APEX 
PA93 chip (Apex Microtechnology Corp, Tucson, AZ) following a circuit provided by the 

25 manufacturer. Preferred amplifiers for the present application would typically meet, or exceed the 
following specifications: ±100V DC in, 100 GQ, input impedance, 20X voltage gain, ±90V out, ±3 
A out, 10 Q, output impedance. 

The majority of current passes through the saline between the electrodes, typically in a 
single eight-well column of the microtiter plate (350) at a time. Excitation light at 400 ± 7.5 nm 

30 illuminates the stained cells from below, and emitted fluorescent light is measured at two 
wavelengths via the detector module (340) blue at 460 +/- 20 nm and orange at 580 +/- 30 nm; (see 
Gonzalez et aL, Drug Discovery Today 4: 431-439, 1999). Once a column of cells has been 
stimulated the computer (300) triggers the motor (360) to move the multiwell plate (350) to a new 
position ready for the next stimulation. 

35 For a typical 96-well multiwell plate, the electrodes are 4 mm wide with a gap (g) of 4 

mm. Stimulation is usually performed in a volume of 100 \xL of physiological saline in the well. 



-26- 



WO 02/08748 PCT/US01/21652 

With this volume of saline, the depth averages approximately 3.0 mm (this depth varies by as 
much as 20% across the well due to the meniscus effect). The electrodes rest approximately 0.5 
mm off the bottom of the wells. The electric field (E) applied across the cells is estimated as the 
voltage across the electrodes (Vq) divided by the electrode gap (g), E = V Q /g . This is an 
5 overestimate of the actual field because of the influence of electrochemical reactions at each 
electrode which consume approximately 1.5 V. In the typical voltage ranges used for stimulation 
(10 to 60 V/cm), this overestimate is on the order of approximately 10%. Accurate measurement 
and calibration of the field can be performed by mapping the electric potential in the well when 
current is passed. 

10 The present invention also includes automated workstations that are programmably 

controlled to minimize processing times at each workstation and that can be integrated to minimize 
the processing time of the liquid samples for electrical stimulation and analysis. 

Typically, a system of the present invention would include one or more of the following: 
A) a storage and retrieval module comprising storage locations for storing a plurality of chemicals 

15 in solution in addressable chemical wells, a chemical well retriever and having programmable 
selection and retrieval of the addressable chemical wells and having a storage capacity for at least 
1 00,000 addressable wells, B) a sample distribution module comprising a liquid handler to aspirate 
or dispense solutions from selected addressable chemical wells, the chemical distribution module 
having programmable selection of, and aspiration from, the selected addressable chemical wells 

20 and programmable dispensation into selected addressable sample wells (including dispensation 
into arrays of addressable wells with different densities of addressable wells per centimeter 
squared), C) a sample transporter to transport the selected addressable chemical wells to the sample 
distribution module and optionally having programmable control of transport of the selected 
addressable chemical wells (including adaptive routing and parallel processing), D) a system for 

25 automated washing, staining, and timed incubation of cells in multiwell plates, E) a system for 
automatically transporting cell plates and test compound plates between the various workstations, 
F) a system for automated electrical stimulation and spectroscopic measurement, and a data 
processing and integration module, G) a master control system which co-ordinates the activities of 
any of the above subsystems. 

30 The storage and retrieval module, the sample distribution module, and the system for 

automated electrical stimulation and spectroscopic measurement are integrated and programmably 
controlled by the data processing and integration module. The storage and retrieval module, the 
sample distribution module, the sample transporter, the system for automated electrical stimulation 
and spectroscopic measurement and the data processing and integration module are operably linked 

35 to facilitate rapid processing of the addressable sample wells. Typically, devices of the invention 



-27- 



WO 02/08748 PCT/US01/21652 

can process at least 100,000 addressable wells in 24 hours. This type of system is described in 
U.S. Patent No. 5,985,214, issued 11/16/99. 
Microfluidic Systems 

The present invention also includes the use of electrodes that have been incorporated into 
5 microfluidic chips and which provide for highly miniaturized electrical stimulation and analysis. 
Such systems include those, for example, described in U.S. Patent No., 5,800,690 issued 
September 1, 1998 to Chow et al, European patent application EP 0 810 438 A2 filed May 5 1997, 
by Pelc et ah and PCT application WO 98/00231 filed 24 June 1997 by Parce et ah These systems 
typically use electrogenic fluid movement to manipulate small fluid volumes within 

10 microcapillaries present on glass or silicon chips. These microfluidic chip based analysis systems 
can provide massively parallel miniaturized analysis. Such systems are preferred systems of 
spectroscopic measurements in some instances that require miniaturized analysis. 

For example, the microfabricated fluorescence-activated cell sorter described by Fu et ah 
(Nature Biotechnology J/7: 1109-11, 1999) could be easily modified to have a pair of electrodes 

15 placed in, or near the optical interrogation region. Using the methods described herein, individual 
cells could be electrically stimulated and individually sorted based on their response to the 
stimulation. This method would greatly simplify the process of obtaining stable clones containing 
the desired expression of channels. In another aspect, screening of test compounds on single cells 
could be performed with a microfluidic device equipped with one or more additional fluid injection 

20 ports and one or more embedded electrical stimulator devices built and operated based on the 
methods described herein. 

V. Electrical Stimulation Methods 

a) Introduction 

Without being bound to any mechanism of action, the present inventors provide the 
25 following description for the effect of electrical stimulation on cellular transmembrane potentials. 

Typical voltage-dependent ion channels have a variety of conducting and non-conducting 
states that are regulated by the local relative transmembrane potential of the cell. By appropriately 
applying external electrical fields to the cells, portions of the cell membrane can be driven to any 
desired transmembrane potential, thereby enabling the regulation of the conduction states of 
30 voltage dependent ion channels present within the cell. If the applied electrical field is 
appropriately varied, it is possible to sample a number of conductance states of most ion channels, 
thereby cycling them through resting, activated, and inactivated states. 

Depending on the ion channel in question, activation of the ion channel can lead to the 
release, or uptake, of ions into the cell that can result in global transmembrane potential changes in 
35 the cell. By applying a repetitive train of electrical stimuli, separated by a time interval smaller 
than the membrane time constant, large sustained membrane voltage changes can be created via a 



-28- 



WO 02/08748 PCT/US01/21652 

stepwise accumulation or loss of ions. This process allows the direct measurement of many ion 
channels and provides a facile method whereby the transmembrane potential of the cell can be 
externally controlled. This approach therefore provides for improved methods of drug discovery 
that are compatible with high throughput screening. 

5 b) Overview of a typical stimulation protocol 

The simulated influence of a typical biphasic electrical stimulation protocol on a cell line 
expressing a voltage activated sodium channel is illustrated, in simplified form, below. The 
following description assumes that the cell line has no significant expression of other ion channels, 
and that the resting transmembrane potential of the cell is above the threshold for inactivation of 

1 0 the sodium channel in question. In FIG. 4, the upper panel shows the time course of the applied 
electrical field (E), the middle panel shows the simulated inward sodium currents (iNa) * n response 
to the applied electrical field, and the lower panel shows the idealized average transmembrane 
potential of the cell (V m ). In this example, the recordings relate to the changes in these parameters 
that a single cell placed in the center of the applied electrical field would be typically expected to 

1 5 experience during an electrical stimulation wave train. 

Referring to the first pulse, establishing the first electrical field causes a potential drop 
across the cell that is maximal, with respect to the resting transmembrane potential of the cell, at 
the edges of the cell closest to the electrodes (see Hibino et aL, Biophysical Journal 64:1789-1800, 
1993; Gross et al. 1986, Biophys. J. 50:339-348). The magnitude of the electric field-induced 

20 transmembrane potential change AV m at a given point of the membrane in an idealized spherical 
cell can be described by the formula (Ehrenberg et al., Biophys. J. 51:833-837, 1987): 



AV m =-fgrEcos&. 



(1) 



In Equation 1,/is a factor dependent upon the conductivity of the membrane, g is a geometric 
25 factor of order 1, r is half the diameter of the cell parallel to the electric field, E is the local 
magnitude of the electric field, and 6 is the angle between the local direction of the field and a line 
drawn from the center of the cell to the point of the surface being considered. For most intact 
mammalian cells, in which the membrane conductivity is very low compared to the conductivity of 
the solution bathing the cells, the factor /« 1. In practice, cells are rarely spherical when attached 
30 to a substrate and an accurate estimate of the actual magnitude of the electrical field induced 
transmembrane potential changes may be empirically determined. 

As a result of the applied electrical field, the membrane on the side nearest to the anode is 
driven negative, while the membrane on the side nearest the cathode is driven positive. In cells in 
which one edge is driven sufficiently negative to locally lower the transmembrane potential below 



-29- 



WO 02/08748 PCT/US01/21652 

the threshold potential for release of inactivation for the ion channel in question, the applied 
electrical field causes the sodium channels located on this edge to enter the resting state. On the 
other side of the cell, the transmembrane potential is driven positive of the resting potential. 
Because the resting transmembrane potential of the cell is assumed to be above the threshold for 
5 inactivation, sodium channels on this side of the cell remain inactivated and do not pass current. If 
the resting transmembrane potential were instead below the inactivation threshold, channels on this 
side of the cell would activate and pass current. 

When the applied field is reversed, the profile of transmembrane potential changes also 
reverses. The transmembrane potential changes induced by the electric field on the patches of 

10 membrane at the extreme edges of the cells switches polarity. The channels on the side that was 
driven negative during the first phase of stimulation are now driven positive. If the stimulation 
parameters are properly chosen, these channels are now driven above the activation potential and 
begin to allow sodium ion influx. This is shown in FIG. 4, as the first smaller peak of sodium 
influx into the cell. The sodium channels rapidly inactivate after a characteristic time. Meanwhile, 

15 on the other side of the cell, the transmembrane potential is driven negative so that the sodium 
channels release from inactivation and move into the resting state. 

When the second stimulus phase ends, all parts of the membrane rapidly return to a new 
average transmembrane potential. If the average transmembrane potential is now above the 
activation potential of the sodium channels, the channels on the side of the cell that was driven 

20 negative during the second phase of stimulation activate and begin to allow sodium ion influx. 
This is shown in FIG. 4, as the second larger peak of sodium influx into the cell. The sodium 
channels rapidly inactivate after a characteristic time. In this case sodium influx is typically larger 
from the second side than the first side, since the driving force for sodium entry is larger when this 
part of the membrane is driven more positive by an electric field. 

25 Each pulse of sodium channel influx raises the average transmembrane potential of the cell 

(FIG. 4, lower panel). This rise in transmembrane potential can be detected by any of the methods 
described herein, but is conveniently measured via fluorescence emission ratio changes of a FRET 
based voltage-sensitive dye. Due to leakage currents present in all cells, this average 
transmembrane potential shift decays exponentially to the original resting transmembrane 

30 potential. The time dependency of this response, the membrane time constant (x m ), depends upon 
the membrane capacitance and membrane resistance, and is highly variable from one cell type to 
another. For example, time constants can vary from 100 |ns to over one second, depending on the 
cell type. Typically the membrane time constant is around 100 ms for most engineered cell lines. 

To provide a net accumulation of sodium influx the stimulus pulse is repeated before the 

35 transmembrane potential has time to decay to the resting transmembrane potential. During 
subsequent rounds of electrical stimulation, positive charge is steadily accumulated into the cell 



-30- 



WO 02/08748 PCT/US01/21652 

raising the average transmembrane potential in an approximately stepwise fashion with each 
repetition of electrical stimulation. After each pulse of electrical stimulation, the magnitude of the 
sodium ion influxes become steadily smaller as the average transmembrane potential approaches 
the sodium ion reversal potential. Eventually an equilibrium transmembrane potential is 
5 established in which leakage of current out of the cell equals the current influx due to electrical 
stimulation. 

c) Adjustable parameters for the stimulus waveforms 

The present invention includes the use of any waveform kernel with any repetition 
procedure capable of selectively activating ion channels in living cells. The kernel is the 

10 repeatable structure that forms the basis of the stimulus train. In FIG. 4, the kernel is a biphasic 
square pulse, but in principle it could be any limited-time wave function. The time duration of the 
kernel sets the maximum rate at which it can be repeated. The repetition procedure dictates how 
and when the kernel is presented to the sample. In FIG* 4, the repetition rate is fixed and continues 
for a total of ten cycles. However the repetition rate need not be fixed. 

1 5 Additionally, the kernel can be changed during the stimulus train, so that each time the 

repetition procedure calls for a stimulus pulse, a different wave function could be used. 
Furthermore, a feedback mechanism could be used to alter the kernel and/or the repetition 
procedure based upon the measured response of the system. 

The use of arbitrary waveform generators to create the stimulus kernels and trains allows 

20 for a virtually unlimited variation in the waveform in order to tune the electrical stimulus to a 
particular cell type or specific ion channel. The pulse train can be readily modulated via the 
variation of a number of separately controllable components. 
1 . The shape of the individual pulses. 

The waveform kernel that is repeated during the stimulus train can be changed with nearly 

25 endless permutations using a arbitrary digital waveform generator, such as Tektronix AFG 310. 
FIG. 5 shows a schematic representation of a biphasic square waveform to illustrate some of the 
variables that can be modulated. In FIG. 5, the pulse train consists of a starting field E\ (400), that 
lasts for a time t\ 9 a rapid increase in potential (410), that takes a time t2, until reaching a first 
stimulating field E2 (420) that lasts for time t3, a rapid decrease in potential (430) that takes time 

30 t4, until reaching a second stimulating field (440), E3 that lasts a time ts, a rapid increase in 
potential (460) that takes time t6, until reaching the finishing field (470), E4 that lasts a time tj 
until the cycle is repeated. The magnitude and polarity of the electrical fields to E4 are 
separately controllable and may be both statically and dynamically varied as described below. The 
times for which the electrical potentials are applied to the cells, times t\ 9 t3, t5, and tj are also 

35 separately controllable and may be both statically and dynamically varied between 0 and 10 s 
during a wave train, as described below. Finally the changes in potential that occur over times t2, 



-31- 



WO 02/08748 PCT/US01/21652 

t4 and tg , may occur over variable time periods between 0 and 100 ms and be either linear or non 
linear to create waveforms of variable shapes. 

Some examples of these types of variation in the waveform are shown in FIG. 6 (a) 
Biphasic waveform, as shown in FIG. 5, repeated at a rate f. (b) A modified biphasic waveform. 
5 A short interval has been added between the stimulation phases of the wave train. This allows 
current to flow through the channels released from inactivation during the first pulse, (c) 
Monophasic waveform. Only channels on the side of the cell facing the anode will be released 
from inactivation. (d) A ramped waveform. The anode-facing channels will be released from 
inactivation by the square wave. The channels will activate and pass current during the ramp. The 

10 ramp allows the channels to open and pass current at more negative local potentials, so that even 
when the cell is near the reversal potential for sodium ions, large currents can still flow. The point 
along the ramp at which the channels will open varies, (e) A biphasic triangular or sawtooth 
waveform. Ramping may allow the voltage-dependent transitions between states to occur more 
uniformly as the global membrane potential changes. Monophasic triangular waveforms are also 

15 possible, (f) A sinusoidal waveform. This type of waveform may reduce electrical noise during 
high frequency stimulation, (g) A short burst of sinusoidal waveforms, (h) Bursts of sinusoidal 
waveforms, each with different fundamental frequency. This type of stimulation may prove useful 
for studying plasticity effects. The first burst(s) are used to train the system or begin a process, 
while the subsequent bursts(s) are used to assay the system. 

20 Variations in waveform shape may be useful in maintaining fixed stimulus conditions 

during the pulse train. For example, the transmembrane potential excursions experienced by a 
highly polarized cell will vary as its average transmembrane potential changes from around —90 
mV at the beginning of the stimulation cycle to around +60 mV after several repetitive stimulation 
cycles. As a consequence, the applied electrical field required to efficiently release an ion channel 

25 from inactivation varies as the average potential of the cell varies during the course of several 
stimulation cycles. To take this effect into consideration it may be useful, under certain 
circumstances, to change the relative balance between the positive (E2) and negative (E3) phases 
of stimulation as the wave-train progresses. 

Some cell lines, for example HEK-293, have a resting average transmembrane potential 

30 below the activation threshold of some voltage-activated sodium channels. In these cells as the 
transmembrane potential rises during stimulation as a result of sodium ion influx, the sodium 
channels can open independently of the applied electrical stimulation. This can be improved by 
using a sloped current pulse (i.e by increasing t2 and t4). Then, the channels can pass current for a 
defined time just above the activation voltage, independent of the average transmembrane potential 

35 of the cell. 



-32- 



WO 02/08748 PCT/US01/21652 

2. The overall amplitude of the individual pulse (E 2 and E 3). 

The magnitude and polarity of the pulse amplitude controls the relative transmembrane 
potential excursions experienced by the cell during a stimulus pulse. Pulse amplitudes can be 
altered for the entire train, or for the individual pulses to accommodate different channels and cell 
5 types, as discussed in more detail below. In general, the magnitudes of E2 and E3 are selected to 
ensure that the ion channel of interest is efficiently activated, and released from inactivation during 
each stimulation cycle, while at the same time not of sufficient magnitude so as to cause 
irreversible electroporation of the cells. Preferred pulse amplitudes for E2 and E3 are typically in 
the range of 5 to 60 V/cm for most ion channels when expressed in non-excitable mammalian cells 

10 with average sizes from 10 to 25 (im, and may vary either positive or negative relative to earth. As 
above, the amplitude of the stimulus can be changed during the pulse train to maintain stable 
stimulus conditions as the average transmembrane potential changes. Preferred pulse amplitudes 
are inversely dependent upon average cell size. So, the technique can also be used on cells which 
are smaller or larger than 1 0 to 25 |am, by altering the pulse amplitude. 

1 5 3. The duration of the individual pulses and t O. 

Many channels require alterations in the transmembrane potential for extended periods of 
time to release them from inactivation, prior to opening. For example, many voltage-dependent 
sodium channels generally need to experience a transmembrane potential below -90 mV for 
several milliseconds before they are released from inactivation. Efficient use of the electrical 

20 stimulation protocol therefore typically requires that the duration of the pulses t3 and t5 are 
sufficient to enable complete, or almost complete, release from inactivation for the ion channel of 
interest. In some cases it may be desirable to tune the magnitude of t3 and t5 to enable the 
selective release from inactivation of one class, but not another class of ion channel in a cell that 
expresses several ion channel types. In other cases it may be desirable to make t3 and t5 very 

25 small to achieve low levels of release from inactivation for the channels. Typically the preferred 
pulse duration is matched to the characteristic time for transitions between the desired voltage- 
dependent states for the ion channel of interest, and these are typically in the range of about 0.1 to 
100 msec for most ion channels. 

To avoid excessive electrolysis of water and consequent gas bubble generation, the 

30 duration of the pulses t3 and t5 should be kept as short as possible, while still achieving the desired 
electrical stimulation. Water electrolysis at a metal/water interface typically occurs when the 
magnitude of the voltage difference between the metal and the water exceeds about 0.8 V. In some 
cases, the stimulus parameters required to produce cellular stimulation also cause water 
electrolysis. Some generation of gas at the electrodes is typically acceptable as long as the charge 

35 per unit area of the electrode/water interface delivered during any single polarity phase of a single 
pulse is less than about 100 jnC/mm^. Exceeding this limit typically causes gas evolution and 



-33- 



WO 02/08748 PCT/US01/21652 

bubble formation that significantly affects field uniformity. The presence of bubbles on the 
electrode surface occludes that part of the electrode, and can cause alterations in the electric field 
uniformity. Generation of large amounts of gas can also cause oxidative damage to the cells and 
the dyes in the well. 

5 In a 96- well plate with 100 jiL of physiological saline with resistivity 70 Q-cm, the 

resistance of the saline between two parallel plate electrodes with a 4 mm gap between them 
inserted into the well to within 0.5 mm of the bottom of the well, is approximately 230 Q.. Each 
electrode has a contact area with the saline of about 24 mm^ Thus, any single-polarity phase of 
the stimulus protocol should not deliver more than about 2.4 mC of charge. A voltage difference 

10 of about 10 V applied between the plates generates an electric field of about 25 V/cm in the saline. 
This voltage will draw about 43 mA of current. Thus for this electrode configuration, a square 
wave, single-polarity pulse should not exceed about 55 milliseconds in duration in order to limit 
the charge to less than 2.4mC. 
4. The gap between successive stimuli (t \ and t j). 

15 Changing the value of t\ and tj globally for the train, or adjusting it for each individual 

pulse during the train, is useful for optimizing the stimulation protocol for specific ion channels. 
Additionally the approach is also useful for determining certain cellular and channel properties 
including the open channel time and the time course of the channel activation and inactivation. 

For example, for assays involving voltage regulated sodium channels, the insertion of a 

20 time delay (t\+tj) between pulses equal to, or less than, the average sodium channel open time 
allows for a quantitative measurement of the inactivation kinetics of the channel. The inactivation 
kinetics are directly related to the average open channel time. Thus, assays using short interpulse 
intervals allows for the detection of compounds whose primary effect is on inactivation kinetics, a 
mechanism which is otherwise inaccessible using high-throughput techniques. 

25 In most cases the time delay between successive stimuli would be less that the membrane 

time constant in order to obtained sustained increases in transmembrane potential. Typically 
optimal frequencies of stimulation if) are within the range x]yfl - / ^ x b~* where xm is the time 
constant for decay of transmembrane potential changes, and xb * s the average channel open time. 
Some channels do not inactivate, and for these cells the stimulation frequency may be determined 

30 empirically. Additionally, the stimulation frequency / cannot exceed the inverse of the time 
duration of the stimulus kernel. 

Additionally, for certain cell types, it may prove desirable to stimulate at a slower rate. 
For example, slower stimulation rates may be preferred for cells with high channel densities, or for 
assays in which higher pharmacological sensitivity is required. Alternatively for these cases, a 

35 monopolar stimulus could be used. This would only release from inactivation the sodium channels 
on one side of the cell, but the maximum frequency of stimulation could be doubled. 



-34- 



WO 02/08748 PCT/US01/21652 

5. The duration of the train of pulses, or number of pulses in the train. 

Cellular and channel properties can be assayed both in dynamic (i.e. rise and fall times, 
alterations in response shape, etc.) and static modes. Both modes require stimulus train durations 
long enough to explore all the events of interest, yet not longer than necessary to complete the 
5 assay. Typical stimulation times comprise 10 msec pulses, at 25 V/cm pulses repeated at a 
frequency of 20 Hz for 3 seconds. Adjusting these parameters allows assay times to be reduced, or 
to explore processes with both fast and slow time scales. 

6. Multiple pulse trains. 

In some cases it is useful to repeat pulse trains, or to perform a measurement on the same 

10 cells with two different pulse trains. One example would be to completely characterize the 
properties of a channel by measuring the response as a function of stimulus frequency and 
duration, using a single plate of cells subjected to multiple stimulus trains. Another example 
would be to examine plasticity of the response (i.e. activity-dependent changes in response). One 
or more stimulus trains would condition the response, while sets of measurement trains before and 

15 after the conditioning would determine the changes due to activity. 

Feedback of stimulus parameters based upon dynamic measurements of the response. 

The present invention can also be used to create a voltage clamp device, by using a 
dynamic feedback loop to maintain the average transmembrane potential at a preset value. By 
measuring the transmembrane potential using a fast fluorescent output as described below, then 

20 changing stimulus parameters to compensate for any changes in transmembrane potential, it is 
possible to dynamically control the transmembrane potential of the cells. The current necessary to 
maintain that potential would then be determined by computer control of the stimulus parameters. 
The use of high frequency stimulation to avoid electrolysis 

During typical stimulation parameters, a peak current of approximately 50 mA passes 

25 through the solution between the electrodes. During this time various electrochemical reactions 
occur which typically generate toxic species to the cells. Preliminary experiments have shown that 
most mammalian cells typically respond normally for approximately two minutes of electrical 
stimulation using stainless steel electrodes. However prolonged stimulation for longer time periods 
appears to lead to a loss in cell health and viability. At sufficiently high pulse frequencies, such 

30 that the metal-saline interface does not reach the potential for electrolysis of water (approximately 
±1V for stainless steel in saline), current can be passed capacitively and no toxic products will be 
generated. In the electrical stimulator shown in FIG. 1, in which each electrode has an area of 
about 24 mm 2 in contact with the saline, the capacitance per electrode is around 1-10 jiF 
(Robinson, 1968, Proc. IEEE 56:1065-1071). At 50 mA, this capacitance charges to 1 V in around 

35 20-200 |is. This is at the lower limit of the useful pulse duration times. 



-35- 



WO 02/08748 PCT/US01/21652 

Alternatively it is possible to perform electrical stimulation without generating electrolytic 
products. Several treatments are available which can increase the capacitance of the metal-saline 
interface by factors of 2-100. These include surface roughening, electroplating with platinum 
black or gold black, and deposition and activation of iridium/iridium oxide, titanium/titanium 
5 nitride, or polypyrrole films. Using stimulation parameters, which avoid irreversible 
electrochemistry, these surface treatments do not degrade when passing current. 
VI. Expression of Ion Channels 
a) Selection of the cell type 

The present invention can be used with any type of cell, including animal cells, plant cells, 
10 insect cells, bacterial cells, yeast and mammalian cells. For screening for human therapeutics 
mammalian cell lines are preferred, such cell lines include tissue culture cell lines that can be 
relatively easily grown, and can be readily transfected with high efficiency. Many tissue cell lines 
are commercially available through the American type culture collection (ATCC) see 
( http://www.atcc.orgX as well as the European collection of cell cultures (ECACC) 
15 ( http://www.camr.org.uk\ 

Additionally in some cases primary cell lines, or tissue slices may also be preferred for 
screening when it is required to express, or measure, the response of the ion channel of interest in 
its native physiological context. This approach may be useful either as a primary or a secondary 
screen to screen for specificity, selectivity or toxicity of candidate therapeutics, and is discussed in 
20 detail in section X. 

For assays performed on cultured cell lines, the main selection criteria are the resting 
transmembrane potential of the cell line, and the presence of endogenously expressed ion channels. 
The selection of appropriate cell lines for specific ion channels of interest are dependent on the 
voltage dependent properties and ion selectivity of the ion channel of interest. These 
25 considerations are reviewed in detail for a number of ion channels in section VIII, Stimulation 
Protocols. 

In some cases it is desirable to use a cell line which has no (or very low) detectable 
endogenous expression of other ion channels. Cells of this type include CHO-K1, CHL, and 
LTK(-) cells. These cells inherently have a resting potential in the range of +10 to -30 mV, which 

30 is above the activation and inactivation thresholds of most voltage-dependent channels. Use of 
these cell lines has the advantage that the ion channel of interest is the major modulator of 
transmembrane potential within the cells so that screening assay data are generally easily and 
unambiguously interpreted. 

In some cases the use of a cell line with no other ion channels may not be practical to 

35 create a workable assay. For example, it may be necessary to maintain a voltage-regulated ion at a 
highly polarized transmembrane potential. In this case it is necessary control the transmembrane 



-36- 



WO 02/08748 PCT/US01/21652 

potential via the expression of a second ion channel. For example to assay a rat brain type Ha 
sodium channel in the resting state requires the transmembrane potential to be maintained below 
the threshold activation potential of the sodium channel, in this case around —60 mV. To achieve 
this it is necessary to either co-express an ion channel, such as a potassium inward rectifier, that 
5 can maintain the resting transmembrane potential of the cell to around -90 mV, or identify a cell 
line that endogenously expresses similar ion channels. Cell types of this type include RBL cells 
andHEK-293 cells. 

In other cases it may be necessary to use the expression of a second ion channel, in 
conjunction with electrical stimulation to drive the cell membrane to a specific transmembrane 

10 potential, to enable the first ion channel of interest to be assayed. Examples of this situation occur 
when assaying non-voltage regulated ion channels such as ligand-gated channels. Co-expression of 
a voltage regulated sodium channel, for example in conjunction with electrical stimulation can be 
used to set the transmembrane potential to transmembrane potentials of between about +10 to +60 
mV. By comparison, co-expression of voltage regulated potassium channels in conjunction with 

15 electrical stimulation can set the transmembrane potential to transmembrane potentials of between 
about —90 to -30 mV. These approaches thus enable the effective manipulation of the 
transmembrane potential over a relatively wide range thereby enabling the analysis of virtually any 
ion channel. 

Typically when using this co-expression approach it is necessary to re-screen any hits 
20 obtained with the cell line co-expressing both ion channels, with the cell line expressing only the 
ion channel used to set the transmembrane potential. This enables drugs that affect this second ion 
channel to be differentiated from those that actually influence the ion channel of interest. 
Alternatively selective toxins such as TTX can be used to selectively inhibit one class of ion 
channel. 

25 b) Transfection of ion channels 

Nucleic acids used to transfect cells with sequences coding for expression of the ion 
channel of interest are typically in the form of an expression vector including expression control 
sequences operatively linked to a nucleotide sequence coding for expression of the channel. As 
used, the term nucleotide sequence coding for expression of a channel refers to a sequence that, 

30 upon transcription and translation of mRNA, produces the channel. This can include sequences 
containing, e.g., introns. As used herein, the term expression control sequences refers to nucleic 
acid sequences that regulate the expression of a nucleic acid sequence to which it is operatively 
linked. Expression control sequences are operatively linked to a nucleic acid sequence when the 
expression control sequences control and regulate the transcription and, as appropriate, translation 

35 of the nucleic acid sequence. Thus, expression control sequences can include appropriate 
promoters, enhancers, transcription terminators, a start codon (i.e., ATG) in front of a protein- 



-37- 



WO 02/08748 PCT/US01/21652 

encoding gene, splicing signals for introns, maintenance of the correct reading frame of that gene 
to permit proper translation of the mRNA, and stop codons. 

Methods which are well known to those skilled in the art can be used to construct expression 
vectors containing the ion channel coding sequence, operatively coupled to appropriate localization 
5 or targeting domains and appropriate transcriptional / translational control signals. For example by 
reference to the sequence accession numbers, or references in Tables 1 to 3, one or ordinary skill in 
the art can identify the sequence of the ion channel of interest. These methods include in vitro 
recombinant DNA techniques, synthetic techniques and in vivo recombination/genetic 
recombination. (See, for example, the techniques described in Maniatis, et ah y Molecular Cloning 
10 A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., 1989). Many commercially available 
expression vectors are available from a variety of sources including Clontech (Palo Alto, CA), 
Stratagene (San Diego, CA) and Invitrogen (San Diego, CA) as well as and many other 
commercial sources. 

A contemplated version of the method is to use inducible controlling nucleotide sequences 
15 to produce a sudden increase in the expression of the ion channel of interest e.g., by inducing 
expression of the channel. Example inducible systems include the tetracycline inducible system 
first described by Bujard and colleagues (Gossen and Bujard (1992) Proc. Natl. Acad. Sci USA 89 
5547-5551, Gossen et al. (1995) Science 268 1766-1769) and described in U.S. Patent No 
5,464,758. 

20 Transformation of a host cell with recombinant DNA may be carried out by conventional 

techniques as are well known to those skilled in the art. Where the host is prokaryotic, such as E. 
colU competent cells that are capable of DNA uptake can be prepared from cells harvested after 
exponential growth phase and subsequently treated by the CaCl2 method by procedures well 
known in the art. Alternatively, MgCl2 or RbCl can be used. Transformation can also be 

25 performed after forming a protoplast of the host cell or by electroporation. 

When the host is a eukaryote, such methods of transfection of DNA as calcium phosphate 
co-precipitates, conventional mechanical procedures such as microinjection, electroporation, 
insertion of a plasmid encased in liposomes, or virus vectors may be used. Eukaryotic cells can 
also be co-transfected with DNA sequences encoding the ion channel, and a second foreign DNA 

30 molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene. 
Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine 
papilloma virus, to transiently infect or transform eukaryotic cells and express the ion channel. 
{Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory, Gluzman ed., 1982). Preferably, a 
eukaryotic host is utilized as the host cell as described herein. 

35 Selection of stable clones will typically be made on the basis of successful expression of 

the ion channel of interest at sufficient level to enable it's facile detection. In many cases this 



-38- 



WO 02/08748 PCT/US01/21652 

analysis will require functional characterization of individual clones to identify those that exhibit 
appropriate electrophysiological characteristics consistent with expression of the clone of interest. 
This analysis can be completed via the use of patch clamping, or via the measurement of 
transmembrane potentials using transmembrane potential sensitive dyes as described below. An 
5 advantage to the use of this latter method is that it is compatible with fluorescence activated cell 
sorting and provides for the rapid analysis of many thousands of individual clones per second. In 
some cases where the sodium channel is electrically silent in the resting cell, confirmation of 
expression can also be readily achieved by immunochemistry using antibodies raised against the 
native ion channel, or a defined epitope introduced in the ion channel via molecular techniques as 

1 0 described above. 

In cases where cells are transfected with a first ion channel of interest, and a second ion 
channel to set the transmembrane potential, optimization of the relative expression of both ion 
channels is important. Typically the optimal relative expression of the two ion channels is 
determined empirically by selecting clones that provide the maximum dynamic range and minimal 

1 5 statistical variation in their response. 

VII . Measurement of Transmembrane potentials 

Transmembrane potential changes and the measurement of specific ion channels 
conductance via the use of the present invention can be detected by use of any of the known means 
of measuring transmembrane potential or ion movement. These methods include, for example, 

20 patch clamping (Hamill et al, Pfluegers Arch. 391:85-100, 1981), FRET based voltage sensors, 
electrochromic transmembrane potential dyes (Cohen et al., Annual Reviews of Neuroscience 1: 
171-82, 1978), transmembrane potential redistribution dyes (Freedman and Laris, Spectroscopic 
membrane probes Ch 16, 1988), extracellular electrodes (Thomas et al., Exp. Cell Res. 74: 61-66, 
1972), field effect transistors (Fromherz et al., Science 252: 1290-1293, 1991) , radioactive flux 

25 assays, ion sensitive fluorescent or luminescent dyes, ion sensitive fluorescent or luminescent 
proteins, the expression of endogenous proteins or the use of reporter genes or molecules. 

Preferred methods of analysis for high throughput screening typically involve the use of 
optical readouts of transmembrane potential, or ion channel conductance. Such methods include 
the use of transmembrane potential or ion sensitive dyes, or molecules, that typically exhibit a 

30 change in their fluorescent or luminescent characteristics as a result of changes in ion channel 
conductance or transmembrane potential. 

A preferred optical method of analysis for use with the present invention has been 
described in U.S. patent No 5,661, 035 issued August 26, 1997). This approach typically comprises 
two reagents that undergo energy transfer to provide a ratiometric fluorescent readout that is 

35 dependent upon the transmembrane potential. Typically the approach uses a voltage sensing 



-39- 



WO 02/08748 PCT/US01/21652 

lipophilic dye and a voltage insensitive fluorophore associated with a cell membrane, (see 

Gonzalez et al. Drug Discovery Today 4:43 1-439, 1999). 

In one embodiment, two dye molecules, a coumarin-linked phospholipid (CC2-DMPE) and 

an oxonol dye such as bis-(l,2-dibutylbarbituric acid) trimethine oxonol [DiSBAC4(3)], are loaded 
5 into the plasma membrane of cells. CC2-DMPE partitions into the outer leaflet of the plasma 

membrane where it acts as a fixed FRET donor to the mobile, voltage sensitive oxonol acceptor. 

Cells with relatively negative potentials inside will push the negatively charged oxonol to the outer 

leaflet of the plasma membrane, resulting in efficient FRET (i.e. quenching of the coumarin donor 

and excitation of the oxonol acceptor). Depolarization results in rapid translocation of the oxonol 
10 to the inner surface of the plasma membrane, decreasing FRET. Because FRET can only occur 

over distances of less than 100 A, excitation of the coumarin results in specific monitoring of 

oxonol movements within the plasma membrane. 

The response times for these assays is readily altered by increasing or decreasing the 

hydrophobicity of the oxonol. For example, the more hydrophobic dibutyl oxonol DiSBAC4(3) 
15 has a time constant of approximately 10 ms, significantly faster than the less hydrophobic diethyl 

oxonol DiSBAC2(3). 

Loading of the dyes is typically achieved at room temperature prior to the start of 
transmembrane potential measurements. Typically cells are loaded sequentially with the coumarin 
lipid followed by the oxonol. Typical loading concentrations for coumarin lipids range from about 

20 4 to 15 |LiM (final concentration) and staining solutions are typically prepared in Hanks Balanced 
salt solution with 10 mM HEPES, 2g/L glucose and about 0.02% Pluronic-127 at a pH of around 
7.2 to 7.4. Loading is usually acceptable after about 30 minutes incubation, after which excess dye 
may be removed if desired. Oxonol dyes are typically loaded at a concentration between 2 and 10 
jlxM for 25 minutes at room temperature, the more hydrophobic DiSBAC4(3) is usually loaded in 

25 the presence of 2-3 pM Pluronic-127. Optimal loading concentrations vary between cell types and 
can be empirically determined by routine experimentation. Typically such optimization 
experiments are conducted by systematically titrating the concentrations of the first reagent, and 
then for each concentration tested, titrating the concentration of the second reagent. In this way it is 
possible to obtain both the optimal loading concentrations for each reagent, and the optimal 

30 relative ratio to achieve a maximal signal to noise ratio. 

In some cases it may be preferred to add, or load one, or more of the FRET reagents with 
one or more light absorbing substances in order to reduce undesired light emission, as for example 
described in commonly owned U.S. Patent Application No. 09/118,497, filed July 17, 1998; U.S. 
Patent Application No. 09/120,516, filed July 21, 1998, and U.S. Patent Application No. 

35 09/122,477 filed July 23, 1998. 



-40- 



WO 02/08748 PCT/US01/21652 

FRET based voltage sensors may also be derived from the use of other membrane targeted 
fluorophores in conjunction with a mobile hydrophobic donor or acceptor. Other such 
compositions are disclosed, for example, in U.S. Patent Application No. 09/459,956, filed 
December 13, 1999. 

5 Suitable instrumentation for measuring transmembrane potential changes via optical 

methods includes microscopes, multiwell plate readers and other instrumentation that is capable of 
rapid, sensitive ratiometric fluorescence detection. A preferred instrument of this type is described 
in U.S. Patent application 09/118,728 filed July 17, 1998. This instrument (the Voltage/Ion Probe 
Reader or VIPR™) is an integrated liquid handler and kinetic fluorescence reader for 96-well and 

10 greater multiwell plates. The VIPR™ reader integrates an eight channel liquid handler, a multiwell 
positioning stage and a fiber-optic illumination and detection system. The system is designed to 
measure fluorescence from a column of eight wells simultaneously before, during and after the 
introduction of liquid sample obtained from another microtiter plate or trough. The VIPR™ reader 
excites and detects emission signals from the bottom of a multiwell plate by employing eight 

15 trifurcated optical bundles (one bundle for each well). One leg of the trifurcated fiber is used as an 
excitation source, the other two legs of the trifurcated fiber being used to detect fluorescence 
emission. A ball lens on the end of the fiber increases the efficiency of light excitation and 
collection. The bifurcated emission fibers allow the reader to detect two emission signals 
simultaneously and are compatible with rapid signals generated by the FRET-based voltage dyes. 

20 Photomultiplier tubes then detect emission fluorescence, enabling sub-second emission ratio 
detection. 

VIII. Stimulation Protocols 

In one aspect, the present invention includes methods for modulating the transmembrane 
potentials of living cells via electrical stimulation, and the use of these methods for assaying the 
25 activity of virtually any ion channel or transporter system. 

a) Measurement of specific channel conductances 

L Assay of sodium channels 

A variety of different isoforms of mammalian voltage dependent sodium channels have 
been identified, and are summarized below in Table 1. These channels can be classified into three 
30 main groups (for review see Goldin, Annals N.Y. Academy of Sciences 868:38-50, 1999). 



35 



-41- 



WO 02/08748 



PCT/US01/21652 





Table 1 






Sodium Channel Sub-type Summary 




Channel Name & 


Sub-type / Alternate names 


Tissue Distribution 


Accession 


Gene Symbol 






Number 


SCN1A (Navl.l) 










Rat I (rat) 


CNS / PNS 


X03638 




HBSCI (human) 


CNS 


X65362 




GPB1 (Guinea pig) 


CNS 


AF003372 


SCN2A (Navl.2) 




■■■•i: ' : .• ' 






Rat II (rat) 


CNS 


X03639 




HBSCII (human) 


CNS 


X65361 




HBA (human) 


CNS 


M94055 


Nav 1.2A 


Rat II A 


CNS 


X61149 


SCN3A (Nav 1.3) 










Rat III (rat) 


CNS 


Y00766 


SCN4A (Navl.4) 










SkMl 5 (lxI (rat) 


skeletal muscle 


M26643 




SkMl ^human^ 


<J JVwlt Id J. lilUOviv 


M81758 


SCN5A (Navl.5) 










QVA/T9 ^roti 


„ 

skeletal muscle / 


M27902 






heart 






xj. i ^xiumdii ^ 


heart 


A/1779 

1V1 / / Z, J *J 


SCN8A rflVavl 6\ 










NaCh6 (rat) 


CNS / PNS 


L39018 




PN4a f rat"! 


CNS / PNS 


AF049239A 




Scn8a (mouse) 


CNS 


U26707 




Scn8a (human) 


CNS 


AF050736 




Cerlll (Guinea pig) 


CNS 


AF003373 


SCN9A (Navl/7) ^ 


















PN1 (rat) 


PNS 


U79568 




HNE-Na (human) 


thyroid 


X82835 




Nas (rabbit) 


Schwann cells 


U35238 


SCN10A NavL8 




Pi- 






SNS (rat) 


PNS 


X92184 




PN3 (rat) 


PNS 


U53833 




SNS (mouse) 


PNS 


Y09108 


8CN6ANav2.1 










Na2.1 (human) 


Heart, uterus muscle 


M91556 


SCN7A Nav2.2 










Na-G (rat) 


astrocytes 


M96578 




SCLll(rat) 


PNS 


Y09164 


Nav2.3 


Na2.3 (mouse) 


Heart, uterus muscle 


L36179 


Nav3.1 


NaN (rat) 


PNS 


AF059030 


SCNlBNapi.l 










P-l (rat) 


CNS 


M91808 




P-l (human) 


CNS 


L10338 


SCN2BNap2.1 










p-2 (rat) 


CNS 


U37026 




p-2 (human) 


CNS 


AF007783 



-42- 



WO 02/08748 PCT/US01/21652 

The voltage-dependent sodium channels in Table 1 vary widely in their voltage 
dependency and inactivation and activation kinetics. Voltage-gated sodium channels have many 
different conformations, which can be classified into three states. (1) The resting state, in which 
the channel is closed and no current can flow. This is the typical state when a sodium channel is 
5 expressed in a cell with a resting transmembrane potential of below about -60 mV. The channel 
can be rapidly driven into the open state by depolarization, usually to a transmembrane potential of 
above about —50 mV. (2) The activated state, in which the channel is open and ions can pass 
through. Because the intracellular concentration of sodium is low in a normal resting cell, while 
the extracellular concentration is high, sodium ions flow into the cell and drive the transmembrane 

10 potential more positive. The open state has a short lifetime, generally on the order of one 
millisecond, after which it passes into the inactivated state. (3) The inactivated state, in which a 
channel has closed and ions can not pass through the channel. The channel cannot be directly 
opened once in the inactivated state. It will first go to the resting state, which occurs if the 
transmembrane potential is held very negative (generally below -80 mV) for several milliseconds. 

15 The time constants and threshold potentials for transitions between these three states vary greatly 
between channel subtypes. 

During these experiments, the response will be compared for cells with active channels, 
and for cells in which the channels are pharmacologically blocked. If a suitable pharmacological 
agent is not available, the blocked state can be emulated with an un-transfected cell line. The 

20 optimal stimulus parameters will yield the smallest coefficient of variation of the difference in 
signals of the two cell populations. 

i) Assays for voltage-dependent sodium channels in an inactivated state 

Preferred cells include those with resting transmembrane potentials above the activation 
threshold for the ion channel of interest, and in which there are no other ion channels expressed. 

25 Cells meeting these criteria include CHL and LTK(-) cells. After choosing a target ion channel, 
cells are transfected and clones are selected as described in section III. Alternatively, a cell line 
that endogenously expresses the channel of interest, and low levels of other channels, could be 
used. For example, the CHO-K1 cell line expresses a voltage-gated sodium channel, and very low 
levels of other ion channels. Cells are plated into multiwell microtiter plates, cultured, and stained 

30 with voltage-sensitive dyes as described in section IV prior to initiating electrical stimulation. 
Initial experiments are typically carried out in a 96-well multiwell plate, with an equal number of 
cells in each well. Generally columns of eight wells are simultaneously stimulated under identical 
conditions to provide statistically significant data on the variation in cellular response. 

An optimal electrical stimulation protocol should hyperpolarize part of the plasma 

35 membrane of the majority of the cells long enough to release the sodium channels from 
inactivation, prior to providing an activating depolarization, without electroporating or killing the 



-43- 



WO 02/08748 PCT/US01/21652 

cells. Typically this requires sustained transmembrane potentials of around -60 to -80 mV for 
periods ranging from about 0.5 to about 20 ms to be created within the cell. 

A preferred stimulation protocol that achieves this effect is biphasic, so that ion channels 
present on both the extreme edges of the cells are released from inactivation as the biphasic 
5 waveform reverses polarity. Typically one would start out with initial conditions using a biphasic 
square wave kernel of 5 msec per phase and an amplitude of 25 V/cm. The kernel would be 
repeated at a regular rate of about 20 Hz for a total train duration of about three seconds. One 
would then optimize the pulse amplitude (up to a maximum of about 60 V/cm), duration (in the 
range of 0.1 to 50 ms), and then frequency (in the range of 0 to 1kHz). If necessary changes in the 

10 pulse shape could also be explored to determine if these resulted in more efficient electrical 
stimulation. The optimal stimulus parameters will yield the maximum cellular stimulation 
(compared to cells with the channel blocked, or not present) with smallest coefficient of variation 
of the signal among the different test wells, at the lowest electric field strength, and at the lowest 
duty cycle for passage of current through the electrodes. After a particular set of parameters is 

15 chosen, a titration of staining concentrations for the voltage sensor dye(s) should be performed as 
described above, to further optimize the signal size and coefficient of variation of the responses. 
These procedures (dye concentrations, electric field strength, and stimulus duration and frequency) 
can be iterated to further optimize the signal. 
ii) Assays for sodium channels normally in the resting state 

20 Preferred cells include those with resting transmembrane potentials below the activation 

threshold for the ion channel of interest, and in which the expression of other ion channels is 
largely confined to a few characterized ion channel types. Cells of this type include HEK-293 and 
RBL cells as well as Fll and HL5 cells. After choosing a target ion channel, cells are transfected 
with the ion channel of interest and clones are selected as described above. Alternatively, as in the 

25 case of Fll and HL5 cells, endogenous sodium channels can be used. After selection and 
characterization, cell clones are plated into multiwell microtiter plates and stained with voltage- 
sensitive dyes as described above. As previously, initial experiments are typically carried out in a 
96-well multiwell plate, with an equal number of cells in each well. Generally columns of eight 
wells are simultaneously stimulated under identical conditions to provide statistically significant 

30 data on the variation in cellular response. 

A number of assay approaches are possible depending on the expression level of the 
sodium channel of interest in the cell. For high levels of voltage-dependent sodium channel 
expression, the sodium current can be large enough to create a large transmembrane potential 
change after a single channel activation/inactivation sequence. In these cases small positive 

35 perturbations in the transmembrane potential created via electrical stimulation can be sufficient to 
activate enough sodium channels that the subsequent sodium ion entry depolarizes the entire cell 



-44- 



WO 02/08748 PCT/US01/21652 

thereby activating all the sodium channels. The stimulus field should typically be applied for a 
time long enough to activate the channels, but not so long as to interfere with the subsequent ion 
flux. After the cell transmembrane potential has re-polarized, the stimulation procedure can be 
repeated. Subsequent stimulation events can be identical to the first, or varied to examine time- 
5 dependent properties of the channels. 

Typically one would start out with initial conditions using a biphasic square wave kernel of 
500 jus per phase and an amplitude of 10 V/cm. One would then optimize the pulse amplitude 
(between 5 and 60 V/cm) and duration (between 0.1 and 1 ms). If necessary changes in the pulse 
shape could also be explored to determine if these resulted in more efficient electrical stimulation. 

10 The optimal stimulus parameters will yield the maximum cellular stimulation with smallest 
coefficient of variation of the signal among the different test wells, at the lowest electric field 
strength, and at the lowest duty cycle for passage of current through the electrodes. After a 
particular set of parameters is chosen, a titration of staining concentrations for the voltage sensor 
dye(s) should be performed as described above, to further optimize the signal size and coefficient 

15 of variation of the responses. These procedures (dye concentrations, electric field strength, and 
stimulus duration and frequency) can be iterated to further optimize the signal. 

Often it will be necessary to use cells whose expression of sodium channels is too low to 
give acceptable signal sizes from single stimuli. It may also be desirable to maintain a large signal 
over an extended period of time. In these cases, the cells can be given pulse trains as described for 

20 channels held above the activation potential. With biphasic stimulus pulses, the sodium channels 
can be activated independent of the starting transmembrane potential. By keeping the inter-pulse 
interval shorter than the membrane time constant, each stimulus will drive current into the cell 
until an equilibrium between inward and outward currents is established. This voltage deviation 
will be maintained as long as the stimulus train continues. 

25 The stimulation protocols in this case are essentially the same as described for cells whose 

resting potential is above the inactivation threshold. In general, a series of initial experiments are 
conducted using a biphasic square wave kernel repeated at a regular rate for a fixed train duration. 
The pulse duration varies from about 1 \is to about 1 s, and more preferably from about 1 00 \xs to 
about 20 ms. The pulse amplitude varies from 0 V/cm to about 60 V/cm, and more preferably 

30 from 10 V/cm to 50 V/cm. The frequency of stimulation varies between 0 Hz (i.e. a single pulse) 
and 1 00 kHz, and more preferably from 0 Hz to about 1 kHz. The pulse train varies between 0 s 
(i.e. a single pulse) and about 100 s, and more preferably between 0 s and 10 s. The optimal 
stimulus parameters will yield the maximum transmembrane potential changes (compared to cells 
with the channel blocked, or not present) and smallest coefficient of variation of the signal among 

35 the test wells, at the lowest electric field strength. After a particular set of parameters is chosen, a 
titration of staining concentrations for the voltage sensor dye(s) is typically performed as described 



-45- 



WO 02/08748 



PCT/US01/21652 



above to further optimize the signal size and coefficient of variation of the responses. These 
procedures (dye concentrations, electric field strength, and stimulus duration and frequency) can be 
iterated to further optimize the signal. 

b) Potassium channels 

5 Voltage-dependent potassium channels repolarize nerve and muscle cells after action 

potential depolarization. They also play important regulatory roles in neural, muscular, secretory, 
and excretory systems. Most cells actively maintain a high intracellular potassium concentration, 
so that the reversal transmembrane potential for potassium is around -90 mV. Potassium typically 
flows out of the cell, so that opening more potassium-selective channels tends to drive the 
10 transmembrane potential more negative, in contrast to sodium channel opening that typically drives 
the transmembrane potential more positive. 

A summary of the numerous potassium sub-types is presented in Table 2 below. 



Table 2 

Potassium Channel Sub-type Summary 


Channel Type 


Sub-type / Alternate names 


Accession 
Number 


Reference 


ATP regulated 






• ; V 




rKirl.l (ROMK1) (rat) 


U12541 


U.S. Patent 5,356,775 




hKirl . 1 (ROMK1 )(human) 




U.S. Patent 5,882,873 




Kirl.2 


U73191 






Kirl.3 


U73193 




I. 


B-cell 




U.S. Patent 5,744,594 


II. 


h3IR 




U.S. Patent 5,917,027 


III. 


HuKATP-1 




EP 0 768 379 Al 


Constitiiiively 
Active . • .. 










Kir2.1(IRKl) 


U12507 


U.S. Patent 5,492,825 
U.S. Patent 5,670,335 




Kir2.2 


X78461 






Kir2.3 


U07364 




G-proteiri 
Regulated 










Kir3.1 (GIK1, KG A) 


U01071 


U.S. Patent 5,728,535 




Kir3.2 


U11859 


U.S. Patent 5,734,021 




Kir3.3 


U11869 


U.S. Patent 5,744,324 




Kir3.4 (CIR) 


X83584 


U.S. Patent 5,747,278 












Kir4.1(BIR10) 


X83585 














Kir5.1(BIR9) 


X83581 














Kir6.1 


D42145 






Kir6.2 


D5081 














Kir7.1 




EP 0 922 763 Al 



-46- 



WO 02/08748 



PCT/US01/21652 



Table 2 

Potassium Channel Sub-type Summary 


Channel Type 


Sub-type / Alternate names 


Accession 
Number 


Reference 


Voltage Regulated 








KCNA1 


hKvl.l (RCK1, RBK1, MBK1, 
MK1, HuKl) 


LO2750 




KCNA2 


Kvl.2 (RBK2, RBK5, NGK1, 






KCNA3 


Kvl.3 (KV3, RGK5, HuKffl, 






KCNA4 


Kvl.4 (RCK4, RHK1, HuKTT) 






TVOTVT A C 


TV- , 1 C / P T/'\n T T T 1 TTT/'1\ 

Kvl.5 (KV1, HPCN1, HK2) 






KLNAo 


TV-. , 1 *r /'T/'t TJ f~~~<T/~ O T TT> TV O "\ 

Kvl.o (KV2, RCK2, HBK2) 






TV p\T a T 


TV.., 1 T /A /f T/" T>TVZT TT«T/ r ^\ 

Kv 1.7 (Mko, RK6, HaKo) 




U.S. Patent 5,559,009 


Jiv2 {iSftab) 








KCJN131 


TV O 1 /T~\T» TT' 1 oi_ 1_\ 

Kv2.1(DRKl, mShab) 


M64228 




TV OT^TD O 


TV, ,0 O //^TATl TV 1 \ 

Kv2.2 (CDRK1) 








K channel 2 




U.S. Patent 5,710,019 


AvJ (Shaw) 








KCNC1 


Kv3.1 (NGK2) 






KCJNC2 


Kv3.2 (RKShlllA) 






KCNC3 


Kv3.3 (KSliIIID) 


X60796 




KCNC4 


Kv3.4 (Raw3) 






Jiv¥ (Shal) 








KCND1 


Kv4.1 (mShal, KShlVA) 


M64226 




KCND2 


Kv4.2 (RK5, Rat Shal l) 






KCND3 


TV . , A "> /TV 0 1~ YT m ~\ 

Kv4.3 (KSnlVB) 
















l^TV »C 1 /TTVO\ 

nKv5.1(IK8) 




WO 99/41372 












Kvo.l (Kl3) 
















TV, T ' r 7 

Kv7 
















Kv8.l 
















Kv9 






Delayed Rectifier 










KvLQTl 


AF000571 


U.S. Patent 5,599,673 












HERG (erg) 


U04270 


PCT WO99/20760 


Calcium regulated 








Ca^ Regulated 
Big 










BKCa (hSLO) 


UH717 






HBKb3 (p-subunit) 




PCT W099/42575 




Maxi-K 




U.S. Patent 5,776,734 
U.S. Patent 5,637,470 | 



-47- 



WO 02/08748 



PCT/US01/21652 



Table 2 

Potassium Channel Sub-type Summary 


Channel Type 


Sub-type / Alternate names 


Accession 
Number 


Reference 


c<2~ Kegulatect- 
small 








KCNN1 


SKCal 


U69883 




KCNN2 


SKCa2 


U69882 




KCNN3 


SKCa3 


U69884 




KCNN4 


SKCa4 (IKCal) 




Muscle Nerve 1999 
22(6) 742-50 












TWIK1 


U33632 





Potassium channels show enormous diversity in terms of activation and inactivation time 
constants and voltage dependencies. In general, voltage-dependent potassium channels show voltage 
dependence similar to sodium channels, being closed at very negative potentials and opening above a 
5 certain threshold. Potassium channels may have multiple resting states, multiple inactivated states, 
and typically a single activated state. Unlike voltage-dependent sodium channels, transitions are 
allowed between most states. These transitions are activation (moving from a resting to the open 
state), deactivation (moving from the open state to a resting state), inactivation (moving from a resting 
or open state to an inactivated state), release from inactivation (moving from an inactivated state to a 

10 resting state), and flickering (moving from an inactivated state to the open state). There is a great 
diversity in the thresholds of the transitions, and in the voltage dependencies of the transition rates. 
Activation time constants range from 0.1 to 1000 ms with threshold activation potentials from -80 to 
+20 mV. Inactivation time constants range from 0.1 to infinity (i.e. no inactivation) with threshold 
potentials from -60 to 0 mV. Time constants for release from inactivation range from 0.5 ms to 100 

15 ms with threshold potentials from -70 to 0 mV. 

Stimulus protocols necessary to obtain measurable channel- dependent signals are 
somewhat dependent upon the specific properties of the channel in question. Because of the 
diversity in parameters in voltage-dependent potassium channels, the optimization of an electrical 
stimulation protocol may take several iterations. 

20 During these experiments, the response will be compared for cells with active channels, 

and for cells in which the channels are pharmacologically blocked. If a suitable pharmacological 
agent is not available, the blocked state can be emulated with an un-transfected cell line. The 
optimal stimulus parameters will yield the smallest coefficient of variation of the difference in 
signals of the two cell populations. 



-48- 



WO 02/08748 PCT/US01/21652 

Assays using direct stimulation of the potassium channel 
Voltage Regulated potassium Channels 

Because potassium channels generate outward currents, activating the channels causes 
negative transmembrane potential changes. Under physiological conditions, the reversal potential 
5 for potassium is around -90 mV. Because cells expressing only a voltage-dependent potassium 
channel generally have resting potentials near the activation threshold, direct stimulation should 
work for those voltage-dependent potassium channels which have activation thresholds above 
about -50 mV. While small negative deflections in the transmembrane potential (less than 40 mV 
change) can be reliably detected using the FRET voltage-sensitive dyes, it is often preferable to 

10 perform high-throughput screens with larger signals. 

Preferred cell types include those cells that express a minimal level of other ion channels, 
such as CHO-K1, CHL, and LTK(-). The transfection and selection of clones expressing ion 
channels of interest will generally be performed as described above for sodium ion channels 
normally in the resting state. Alternatively, a cell line which endogenously expresses the channel 

15 of interest could be used. The labeling and measurement of cells with transmembrane potential 
dyes will generally be performed as described for sodium ion channels normally in the resting 
state. 

The stimulation protocol will advantageously depolarize part of the plasma membrane long 
enough to activate the voltage-dependent potassium channels. Unlike the case for voltage- 

20 dependent sodium channels, voltage-dependent potassium channels will typically pass current 
during the depolarizing phase of the stimulus pulse. On the side of the cell where the 
transmembrane potential is driven in a negative direction, the potassium channels release from 
inactivation (if the channel in question experiences voltage-dependent inactivation). On the side of 
the cell where the transmembrane potential is driven in a positive direction, potassium channels 

25 activate and pass outward current. Thus, the stimulus pulse duration should not greatly exceed the 
inactivation time. The potassium current tends to drive the average transmembrane potential 
negative of the resting potential. After the stimulus pulse, the transmembrane potential will 
exponentially relax to the resting potential. By repeating the stimulus after a time shorter than the 
membrane time constant, the average cell membrane can be driven further negative. Using a train 

30 of stimuli, a large and sustained signal can be obtained. 

A preferred stimulation protocol that achieves this effect is biphasic, so that ion channels 
present on both the extreme edges of the cells can participate in enabling potassium ion movement. 
Typically one would start out with initial conditions using a biphasic square wave kernel of 5 msec 
per phase and an amplitude of 25 V/cm. The kernel would be repeated at a regular rate of about 20 

35 Hz for a total train duration of about three seconds. One would then optimize the pulse amplitude, 
duration, and then frequency. If necessary changes in the pulse shape could also be explored to 



-49- 



WO 02/08748 PCT/US01/21652 

determine if these resulted in more efficient electrical stimulation. The optimal stimulus 
parameters will yield the maximum average transmembrane potential change (compared to cells 
with the channel blocked, or not present) with smallest coefficient of variation of the signal among 
the different test wells, at the lowest electric field strength, and at the lowest duty cycle for passage 
5 of current through the electrodes. After a particular set of parameters is chosen, a titration of 
staining concentrations for the voltage sensor dye(s) should be performed as described above, to 
further optimize the signal size and coefficient of variation of the responses. These procedures 
(dye concentrations, electric field strength, and stimulus duration and frequency) can be iterated to 
further optimize the signal. 

10 2) Inward-rectifier potassium channels 

Contrary to its name, the function of the inward rectifier channel is not to allow potassium 
into the cell. Inward flow of potassium can only occur (1) when the transmembrane potential falls 
below the potassium equilibrium potentials, or (2) if the extracellular potassium concentration 
rises. Neither situation normally occurs, because (1) under normal physiological conditions, since 

15 potassium is the ion with the most negative reversal potential, no ionic current can drive the 
potential more negative than the potassium reversal potential, and (2) except under pathological 
conditions, the extracellular potassium concentration is tightly controlled. However, using 
electrical stimulation, parts of the cell membrane can be driven below Vk ? promoting potassium 
ion entry into the cell. This will cause a net positive transmembrane potential change and can be 

20 detected as a positive signal. To develop and optimize an assay for blockers of the inward rectifier, 
one could therefore follow the following procedure. 

Preferred cell types include those cells that express a minimal level of other ion channels, 
such as CHO-K1, CHL, and LTK(-). The transfection and selection of clones expressing ion 
channels of interest will generally be performed as described above for sodium ion channels 

25 normally in the resting state. Alternatively, a cell line which endogenously expresses the channel 
of interest could be used. The labeling and measurement of cells with transmembrane potential 
dyes will generally be performed as described for sodium ion channels normally in the resting 
state. 

A preferred stimulation protocol uses a biphasic kernel, so that ion channels present on 
30 both the extreme edges of the cells participate. Typically one would start out with initial conditions 
using a biphasic square wave kernel of 5 msec per phase and an amplitude of 25 V/cm. The kernel 
would be repeated at a regular rate of about 20 Hz for a total train duration of about three seconds. 
One would then optimize the pulse amplitude, duration, and then frequency. If necessary changes 
in the pulse shape could also be explored to determine if these resulted in more efficient electrical 
35 stimulation. The optimal stimulus parameters will yield the maximum cellular stimulation 
(compared to cells with the channel blocked, or not present) with the smallest coefficient of 



-50- 



WO 02/08748 PCT/US01/21652 

variation of the signal among the different test wells, at the lowest electric field strength, and at the 
lowest duty cycle for passage of current through the electrodes. After a particular set of parameters 
is chosen, a titration of staining concentrations for the voltage sensor dye(s) should be performed 
as described above, to further optimize the signal size and coefficient of variation of the responses. 
5 These procedures (dye concentrations, electric field strength, and stimulus duration and frequency) 
can be iterated to further optimize the signal. 

iii) Assays using a voltage-dependent sodium counter-channel 

This method involves the use of a cell line expressing the voltage-dependent potassium 
channel of interest and which also expresses a voltage-dependent sodium channel. In this method 

1 0 the approach is to use electrical stimulation protocols designed to specifically activate the voltage 
dependent sodium channel. In this case electrical stimulation causes sodium ions to enter the cell, 
causing a positive voltage change. The presence of the potassium channel of interest will tend to 
suppress the positive response of the sodium channel by allowing potassium ions to leave the cell. 
The assay takes advantage of the absence of outward current when a test chemical blocks the 

15 potassium channel, thereby restoring the large positive voltage response normally induced by 
activation of the sodium channels. The optimization of the balance of currents is important in this 
method to ensure that the assay is sensitive to potassium channel blockade. If the sodium current is 
too small relative to the potassium current, the dose-response curve for the potassium channel 
blocker will be shifted towards higher concentrations. For example, in the extreme case where the 

20 potassium current is 100 times larger than the sodium current, 99% of the potassium channels 
would have to be blocked in order to get a 50% response from the sodium channels. 

Because this method involves driving a voltage-dependent sodium channel with repetitive 
pulses, the protocol development is essentially the same as described above for voltage-activated 
sodium channels in an inactivated state. Typically one would start out with initial conditions using 
, 25 a biphasic square wave kernel of 5 msec per phase and an amplitude of 25 V/cm. The kernel 
would be repeated at a regular rate of about 20 Hz for a total train duration of about three seconds. 
One would then optimize the pulse amplitude, duration, and then frequency. If necessary changes 
in the pulse shape could also be explored to determine if these resulted in more efficient electrical 
stimulation. The optimal stimulus parameters will yield the maximum cellular stimulation 

30 (compared to cells with the channel blocked, or not present) with smallest coefficient of variation 
of the signal among the different test wells, at the lowest electric field strength, and at the lowest 
duty cycle for passage of current through the electrodes. After a particular set of parameters is 
chosen, a titration of staining concentrations for the voltage sensor dye(s) should be performed as 
described above, to further optimize the signal size and coefficient of variation of the responses. 

35 These procedures (dye concentrations, electric field strength, and stimulus duration and frequency) 
can be iterated to further optimize the signal. 



-51- 



WO 02/08748 PCT/US01/21652 

In this assay format, there will ideally be no (or a very small) response to stimulation in the 
absence of channel block, because the potassium current will counteract the sodium current. 
Therefore, to optimize the stimulus conditions, it will be necessary to compare responses with and 
without the activity of the potassium channel. Ideally, this will be accomplished using a selective 
5 blocker of the potassium channel. In those cases where such a blocker is yet unknown, it will be 
possible to use the cell line containing only the sodium counter-channel. 

Because this assay format involves two ion channels, modulators of either channel will 
affect the voltage response. In this case, a hit (a blocker of the potassium channel) will restore the 
voltage response. The screening format automatically ignores compounds which block only the 

10 sodium channel. However, stimulation of the cells in the presence of compounds which block both 
channels will also result in no voltage deflection, suggesting that the compound is inactive. 
Because compounds of this type may be of interest, a method to unmask them is also available. By 
performing the identical compound screen using the parent cell line, which contains the sodium 
channel but not the potassium channel, blockers of the sodium channel can be found. Compounds 

15 which are found to block the sodium channel can then be tested separately to find if they have 
activity against the potassium channel. 

c) Assay of calcium channels 

Calcium channels are generally found in many cells where, among other functions, they 
play important roles in signal transduction. In excitable cells, intracellular calcium supplies a 

20 maintained inward current for long depolarizing responses and serves as the link between 
depolarization and other intracellular signal transduction mechanisms. Like voltage-gated sodium 
channels, voltage-gated calcium channels have multiple resting, activated, and inactivated states. 

Multiple types of calcium channels have been identified in mammalian cells from various 
tissues, including skeletal muscle, cardiac muscle, lung, smooth muscle and brain, [see, e.g., Bean, 

25 B. P. (1989) Ann. Rev. Physiol. 51:367-384 and Hess, P. (1990) Ann. Rev. Neurosci. 56:337]. The 
different types of calcium channels have been broadly categorized into four classes, L-, T-, N-, and 
P-type, distinguished by current kinetics, holding potential sensitivity and sensitivity to calcium 
channel agonists and antagonists. Four subtypes of neuronal voltage-dependent calcium channels 
have been proposed (Swandulla, D. et al., Trends in Neuroscience 14:46, 1991). 

30 The cDNA and corresponding amino acid sequences of the al, a2, P and y subunits of the 

rabbit skeletal muscle calcium channel have been determined [see, Tanabe et al. (1987) Nature 
328:313-318; Ruth et al. (1989) Science 245:1115-1118; and U.S. Pat. No. 5386,025]. In addition, 
the cDNA and corresponding amino acid sequences of al subunits of rabbit cardiac muscle 
[Mikami, A. et al. (1989) Nature 340:230-233] and lung [Biel, M. (1990) FEBS Letters 269:409- 

35 412] calcium channels have been determined. In addition, cDNA clones encoding a rabbit brain 



-52- 



WO 02/08748 PCT/US01/21652 

calcium channel (designated the BI channel) have been isolated [Mori, Y. et al. (1991) Nature 
350:398-4021, 

Partial cDNA clones encoding portions of several different subtypes, referred to as rat 
brain class A, B, C and D, of the calcium channel al subunit have been isolated from rat brain 
5 cDNA libraries [Snutch, T. et al. (1990) Proc. Natl. Acad. Sci. USA 87:3391-3395]. More recently 
full-length rat brain class A [Starr, T. et al. (1991) Proc. Natl. Acad. Sci. USA 88:5621-5625] and 
class C [Snutch, T. et al. (1991) Neuron 7:45-57] cDNA clones have been isolated. Although the 
amino acid sequence encoded by the rat brain class C DNA is approximately 95% identical to that 
encoded by the rabbit cardiac muscle calcium channel al subunit-encoding DNA, the amino acid 

10 sequence encoded by the rat brain class A DNA shares only 33% sequence identity with the amino 
acid sequence encoded by the rabbit skeletal or cardiac muscle al subunit-encoding DNA. A 
cDNA clone encoding another rat brain calcium channel al subunit has also been obtained [Hui, 
A. et al. (1991) Neuron 7:35-44]. The amino acid sequence encoded by this clone is approximately 
70% homologous to the proteins encoded by the rabbit skeletal and cardiac muscle calcium 

15 channel DNA. A cDNA clone closely related to the rat brain class C al subunit-encoding cDNA 
and sequences of partial cDNA clones closely related to other partial cDNA clones encoding 
apparently different calcium channel al subunits have also been isolated [see Snutch, T. et al. 
(1991) Neuron 7:45-57; Perez-Reyes, E. et al. (1990) J. Biol. Chem. 265:20430; and Hui, A. et al. 
(1991) Neuron 7:35-44]. 

20 For known calcium channels that have been characterized, activation time constants range 

from 0.1 to 10 ms with threshold potentials from -80 to -20 mV. Inactivation time constants range 
from 0.1 to oo (i.e. no inactivation) with threshold potentials from -60 to -20 mV. Time constants 
for release from inactivation range from 0.5 ms to 100 ms with threshold potentials from -70 to -40 
mV. 

25 Choice of cell line and induction of voltage-dependent calcium currents are performed 

using the general guidelines and approaches discussed above for sodium channels. 

Preferred cell types include those cells that express a minimal level of other ion channels, 
such as CHO-K1, CHL, and LTK(-). The transfection and selection of clones expressing ion 
channels of interest will generally be performed as described above for sodium ion channels 

30 normally in the resting state. Alternatively, a cell line which endogenously expresses the channel 
of interest could be used. The labeling and measurement of cells with transmembrane potential 
dyes will generally be performed as described for sodium ion channels normally in the resting 
state. Alternatively, the cells can be loaded with calcium-sensitive fluorescent dyes such as 
Calcium Green, fluo3-AM, or indo-1. 

35 In cells with low background currents, strong inward calcium currents can be generated by 

driving portions of the membrane negative enough to release the channels from inactivation. Then 



-53- 



WO 02/08748 PCT/US01/21652 

by reversing or releasing the external electric field, the channels are exposed to potentials which 
activate the channels and permit calcium current to flow into the cell. The reversal potential for 
calcium in most cells is generally +60 to +100 mV, so large voltage changes due to calcium influx 
are possible. We can use either membrane-bound voltage-sensitive dyes or intracellular calcium 
5 dyes to monitor the activity of the cells. Due to the similarity in properties of calcium and sodium 
channels, the same general assay optimization procedures outlined above for sodium channels will 
apply to calcium channels. 

Typically one would start out with initial conditions using a biphasic square wave kernel of 
5 msec per phase and an amplitude of 25 V/cm. The kernel would be repeated at a regular rate of 

10 about 20 Hz for a total train duration of about three seconds. One would then optimize the pulse 
amplitude, duration, and then frequency. If necessary changes in the pulse shape could also be 
explored to determine if these resulted in more efficient electrical stimulation. The optimal 
stimulus parameters will yield the maximum cellular stimulation (compared to cells with the 
channel blocked, or not present) with the smallest coefficient of variation of the signal among the 

15 different test wells, at the lowest electric field strength, and at the lowest duty cycle for passage of 
current through the electrodes. After a particular set of parameters is chosen, a titration of staining 
concentrations for the voltage sensor dye(s) should be performed as described above, to further 
optimize the signal size and coefficient of variation of the responses. These procedures (dye 
concentrations, electric field strength, and stimulus duration and frequency) can be iterated to 

20 further optimize the signal. 

During these experiments, the response will be compared for cells with active channels, 
and for cells in which the channels are pharmacologically blocked. If a suitable pharmacological 
agent is not available, the blocked state can be emulated with an un-transfected cell line. The 
optimal stimulus parameters will yield the smallest coefficient of variation of the difference in 

25 signals of the two cell populations. 

d) Assay of voltage-dependent chloride channels 

Chloride channels are found in the plasma membranes of virtually every cell in the body. 
Chloride channels mediate a variety of cellular functions including regulation of transmembrane 
potentials and absorption and secretion of ions across epithelial membranes. When present in 

30 intracellular membranes of the Golgi apparatus and endocytic vesicles, chloride channels also 
regulate organelle pH. For a review, see Greger, R. (1988) Annu. Rev. Physiol. 50:1 1 1-122. 

Three distinct classes of chloride channels are apparent based on their type of regulation 
and structural conformation, Table 3. The first class includes the GAB A and Glycine receptor 
super families, the second class includes the CFTR (Cystic fibrosis Transmembrane Conductance 

35 Regulator) and the third class includes the voltage regulated chloride channels. 



-54- 



WO 02/08748 



PCT/US01/21652 



Table 3 

Chloride Channel Sub-type Summary 


Channel Type 


Sub-type 


Tissue 
Distribution 


Reference 


Ligand gated 










GABAa 
Receptor family 


CNS & PNS 


Synapse 21, 189-274 (1995) 




Glycine 

Receptor family 


CNS & PNS 


Trends Neurosci. U, 458-461 (1991) 


cAMP regulated 










CFTR 


Epithelial tissues 


Science 245, 1066-1073 (1989) 


Voltage 
regulated 






'I 




ClC-1 


Skeletal Muscle 


Nature 354, 301-304 (1991) 




ClC-2 


Ubiquitous 


Nature 356, 57-60(1992) 




CIC-Ka 


Kidney 


J. Biol. Chem. 268, 3821-3824 (1993) 




CIC-Kb 


Kidney 


P.N.A.S. 91, 6943-6947 (1994) 




ClC-3 


Broad, e.g. 
Kidney & Brain 


Neuron 12, 597-604 (1994) 




ClC-4 


Broad, e.g. 
Kidney & Brain 


Hum. Mol. Genet. 3 547-552 (1994) 




ClC-5 


Mainly Kidney 


J. Biol. Chem. 270, 31172-31177 (1995) 




ClC-6 


Ubiquitous 


FEBS. Lett. 377, 15-20(1995) 




ClC-7 


Ubiquitous 


FEBS. Lett. 377. 15-20(1995) 



In contrast to ions like sodium and especially calcium, the electrochemical gradient of 
chloride across the plasma membrane is generally not far from equilibrium. Thus, at the resting 
potential of cells, the opening of chloride channels will not lead to large excursions of the plasma 
5 membrane voltage or dramatic changes in intracellular chloride concentrations. Because electrical 
stimulation typically generates symmetrical voltage changes across the cell membrane, no net 
chloride flux can be generated unless the conductivity of the channel is non-linear. For a linear 
leak conductance, a uniform electric field will drive chloride into the cell on one side and out of the 
cell on the other side. 

10 Direct electrical stimulation of chloride channels which have non-linear conductance 

curves (rectifiers) or voltage-activated gating can generate net ion fluxes, which in turn will cause 
detectable transmembrane potential changes. Depending upon the voltage dependence of the 
conductance and gating, the transmembrane potential change can be either positive or negative. 
For typical chloride channels (that activate at elevated potentials and close at more negative 

15 potentials) and for outward rectifiers, chloride will flow into the cell and drive the transmembrane 
potential negative. For inward rectifiers, chloride will be driven out of the cell and the 
transmembrane potential will be driven positive. 



-55- 



WO 02/08748 PCT/US01/21652 

Due to the small difference between the chloride reversal potential and the resting 
transmembrane potential, direct stimulation of a voltage-gated chloride channel may result in 
insufficient transmembrane potential changes. Assays for these ion channels can then be 
developed using co-expression and electrical stimulation of a sodium or potassium counter-channel 
5 in order to produce an inward or outward current. Presence or absence of the chloride current can 
then be determined by the absence or presence of a transmembrane potential change when the 
counter-channel is electrically stimulated. 

Preferred cell types include those cells that express a minimal level of other ion channels, 
such as CHO-K1, CHL, and LTK (-). The transfection and selection of clones expressing ion 

10 channels of interest will generally be performed as described above for sodium ion channels 
normally in the resting state. Alternatively, a cell line which endogenously expresses the channel 
of interest (or the counter-channel) could be used. The labeling and measurement of cells with 
transmembrane potential dyes will generally be performed as described for sodium ion channels 
normally in the resting state. 

15 Typically one would start out with initial conditions using a biphasic square wave kernel of 

5 msec per phase and amplitude of 25 V/cm. The kernel would be repeated at a regular rate of 
about 20 Hz for a total train duration of about three seconds. One would then optimize the pulse 
amplitude, duration, and then frequency. If necessary changes in the pulse shape could also be 
explored to determine if these resulted in more efficient electrical stimulation. The optimal 

20 stimulus parameters will yield the maximum cellular stimulation (compared to cells with the 
channel blocked, or not present) with smallest coefficient of variation of the signal among the 
different test wells, at the lowest electric field strength, and at the lowest duty cycle for passage of 
current through the electrodes. After a particular set of parameters is chosen, a titration of staining 
concentrations for the voltage sensor dye(s) should be performed as described above, to further 

25 optimize the signal size and coefficient of variation of the responses. These procedures (dye 
concentrations, electric field strength, and stimulus duration and frequency) can be iterated to 
further optimize the signal. 

During these experiments, the response will be compared for cells with active channels, 
and for cells in which the channels are pharmacologically blocked. If a suitable pharmacological 

30 agent is not available, the blocked state can be emulated with an un-transfected cell line. The 
optimal stimulus parameters will yield the smallest coefficient of variation of the difference in 
signals of the two cell populations. 

e) Assay of ligand dependent channels 

The ligand-dependent ion channel family is large and diverse. Ligand-dependent ion 

35 channels open in response to the binding of specific molecules. They typically mediate fast 
synaptic transmission between neurons, and from neurons to muscle cells. They also mediate slow 



-56- 



WO 02/08748 PCT/US01/21652 

synaptic transmission and control a variety of regulatory mechanisms. Ligand-gated ion channels 
are generally only charge-selective; that is, they permit the flow of a range of either anions or 
cations but have little specificity. They have enormous variation in their activation, deactivation, 
and desensitization kinetics, all of which can vary from submillisecond to second time constants. 
5 When the ligand binds to the receptor of the channel, the channel undergoes one or more 

conformational changes to activate the channel. If the ligand is removed from the bathing saline, 
the bound ligands dissociate and the channel closes. If the ligand remains in the bathing saline, 
some channels desensitize by retaining the ligand but moving into a different conformational state 
in which the channel is closed. Equilibrium distributions between the activated, deactivated, and 

10 desensitized states vary greatly among channels. 

In current assay formats, the transmembrane potential of the cells is monitored during an 
addition of ligand. The sudden increase in conductance when the channel opens drives the 
transmembrane potential towards a new reversal potential. Unfortunately, for many ligand-gated 
channels, the new reversal potential is usually within 15 mV of the resting potential. This small 

15 change is sufficient to use for signaling within cells, but it makes pharmacological assays difficult. 

In an electrical stimulation assay for ligand-gated ion channels, one approach is to co- 
express a voltage-gated sodium counter channel with the ligand gated ion channel of interest. This 
approach allows us to modulate the transmembrane potential via electrical stimulation. If the test 
compounds are added to the cells during or prior to electrical stimulation, the method enables an 

20 analysis of whether the ligand gated channel is open or closed. If the ligand-gated channels are 
open, the high resting conductance of the cell will suppress the voltage response to electrical 
stimulation. If, however, the ligand-gated channels are blocked, the cells will have a large 
response to electrical stimulation. The large amount of flexibility in electrical stimulation 
parameters should allow us to assay for a large range in resting conductances. This is important in 

25 the case of ligand-gated channels, because the resting conductance in the presence of ligand is very 
sensitive to the equilibrium desensitization. Accounting for desensitization and variations in 
channel expression, we may have resting membrane resistances ranging anywhere from 10 to 
10 GQ. With rat brain type Ha sodium channels as the counter channel, we can cover this entire 
range. It should also be possible to screen for both agonists and antagonists. By choosing 

30 stimulation parameters such that the response is half-size, agonists will reduce the response while 
antagonists will increase it. Better screening windows may be obtained by stimulating at higher 
(agonist assay) or lower (antagonist assay) frequencies. Note that modulators of the channel 
conductance, open time, desensitization, and deactivation will all be detected. 

Preferred cell types include those cells that express a minimal level of other ion channels, 

35 such as CHO-K1, CHL, and LTK (-). The transfection and selection of clones expressing ion 
channels of interest will generally be performed as described above for sodium ion channels 



-57- 



WO 02/08748 PCT/US01/21652 

normally in the resting state. Alternatively, a cell line which endogenously expresses the channel 
of interest (or the counter-channel) could be used. The labeling and measurement of cells with 
transmembrane potential dyes will generally be performed as described for sodium ion channels 
normally in the resting state. 
5 Typically one would start out with initial conditions using a biphasic square wave kernel of 

5 msec per phase and amplitude of 25 V/cm. The kernel would be repeated at a regular rate of 
about 20 Hz for a total train duration of about three seconds. One would then optimize the pulse 
amplitude, duration, and then frequency. If necessary changes in the pulse shape could also be 
explored to determine if these resulted in more efficient electrical stimulation. The optimal 

10 stimulus parameters will yield the maximum cellular stimulation (compared to cells with the 
ligand-gated channel blocked, or not present) with smallest coefficient of variation of the signal 
among the different test wells, at the lowest electric field strength, and at the lowest duty cycle for 
passage of current through the electrodes. After a particular set of parameters is chosen, a titration 
of staining concentrations for the voltage sensor dye(s) should be performed as described above, to 

15 further optimize the signal size and coefficient of variation of the responses. These procedures 
(dye concentrations, electric field strength, and stimulus duration and frequency) can be iterated to 
further optimize the signal. 

During these experiments, the response will be compared for cells with active channels, 
and for cells in which the channels are pharmacologically blocked. If a suitable pharmacological 

20 agent is not available, the blocked state can be emulated with an un-transfected cell line. The 
optimal stimulus parameters will yield the smallest coefficient of variation of the difference in 
signals of the two cell populations. 

f) Assay of passive channels 

Many channels have slow or no voltage-activated conductance changes. Prime examples 

25 are the some of the channels implicated in cystic fibrosis, particularly the cystic fibrosis 
transmembrane regulator (CFTR, a chloride channel), the epithelial sodium channel (ENaC) and 4 
TM potassium channel family members (Wang et al. Ann. N. Y. Acad. Sci. 868: 286-303, 1999). 
A small molecule which acts as an agonist for either of these channels would be a candidate for a 
drug which alleviates cystic fibrosis. Currently, there is no convenient workable high throughput 

30 screening method for channels of this type. 

The proposed assay format for ion channel targets of this type involves a cell expressing 
the leak channel of interest in a cell which also expresses a voltage-dependent sodium channel. 
The channel of interest is cloned into a cell with a voltage-dependent sodium channel. The 
presence of the passive current will suppress the positive response of the sodium channel when the 

35 cells are stimulated. Blocking the passive channel will restore the large positive voltage response. 
Optimization of the balance of currents will be important in this method. Wild-type CHO cell may 



-58- 



WO 02/08748 PCT/US01/21652 

be useful for this purpose, although a cell with larger sodium currents (either endogenous or 
engineered) would be preferable. If the sodium current is too small relative to the potassium 
current, the dose-response curve for the passive channel blocker will be shifted towards higher 
concentrations. For example, in the extreme case where the passive current is 100 times larger 
5 than the sodium current, 99% of the passive channels would have to be blocked in order to get a 
50% response from the sodium channels. 

Preferred cell types include those cells that express a minimal level of other ion channels, 
such as CHOK1, CHL, and LTK (-). The transfection and selection of clones expressing ion 
channels of interest will generally be performed as described above for sodium ion channels 

10 normally in the resting state. Alternatively, a cell line which endogenously expresses the channel 
of interest (or the counter-channel) could be used. The labeling and measurement of cells with 
transmembrane potential dyes will generally be performed as described for sodium ion channels 
normally in the resting state. 

A preferred stimulation protocol uses a biphasic kernel. In general, a series of initial 

15 experiments are conducted using a biphasic square wave kernel repeated at a regular rate for a 
fixed train duration. The pulse duration varies from about 1 jus to about 1 s, and more preferably 
from about 100 \is to about 20 ms. The pulse amplitude varies from 0 V/cm to about 60 V/cm, and 
more preferably from 10 V/cm to 50 V/cm. The frequency of stimulation varies between 0 Hz (i.e. 
a single pulse) and 100 kHz, and more preferably from 0 Hz to about 1 kHz. The pulse train varies 

20 between 0 s (i.e. a single pulse) and about 100 s, and more preferably between 0 s and 10 s. 

Typically one would start out with initial conditions using a biphasic square wave kernel of 
5 msec per phase and an amplitude of 25 V/cm. The kernel would be repeated at a regular rate of 
about 20 Hz for a total train duration of about three seconds. One would then optimize the pulse 
amplitude, duration, and then frequency. If necessary changes in the pulse shape could also be 

25 explored to determine if these resulted in more efficient electrical stimulation. The optimal 
stimulus parameters will yield the maximum cellular stimulation (compared to cells with the 
ligand-dependent channel blocked, or not present) with smallest coefficient of variation of the 
signal among the different test wells, at the lowest electric field strength, and at the lowest duty 
cycle for passage of current through the electrodes. After a particular set of parameters is chosen, a 

30 titration of staining concentrations for the voltage sensor dye(s) should be performed as described 
above, to further optimize the signal size and coefficient of variation of the responses. These 
procedures (dye concentrations, electric field strength, and stimulus duration and frequency) can be 
iterated to further optimize the signal. 

It should be possible to screen for both agonists and antagonists. By choosing stimulation 

35 parameters such that the response is half-maximal, agonists will reduce the response while 



-59- 



WO 02/08748 PCT/US01/21652 

antagonists will increase it. Better screening windows may be obtained by stimulating at higher 
(agonist assay) or lower (antagonist assay) frequencies. 

During these experiments, the response will be compared for cells with active channels, 
and for cells in which the channels are pharmacologically blocked. If a suitable pharmacological 
5 agent is not available, the blocked state can be emulated with an un-transfected cell line. The 
optimal stimulus parameters will yield the smallest coefficient of variation of the difference in 
signals of the two cell populations. 

The present invention also includes methods for the quantitative determination of cellular 
and ion channel parameters, and for the quantification of the pharmacological effects of test 
10 compounds on these parameters using electrical stimulation. 

b) Quantitative measurements of membrane resistances 

After the electrical stimulus ends, the cell transmembrane potential relaxes to a new resting 
potential. In the case of voltage-dependent channel assays, the channels will generally close or 
inactivate, and the final resting equilibrium potential will be the same as before the stimulus. In 

15 most cases, the charge built up on the membrane capacitance will dissipate exponentially through 
the membrane resistance. The membrane time constant is simply the product of the membrane 
capacitance and the membrane resistance, x m =R m C m . It can be readily determined by measuring 
the membrane capacitance and the membrane time constant. 

The average membrane capacitance for cells commonly used in these assays is 

20 independent of the exogenous channel, and can easily be measured by patch clamp methods. The 
membrane time constant can be readily measured by measuring the rate of decay of the 
transmembrane potential and fitting this data to an exponential decay function. Thus by dividing 
the membrane time constant by the average membrane capacitance for the given cell type, we can 
quantitatively determine the resting or leak membrane resistance. 

25 A similar analysis can be made to quantitatively measure the membrane resistance while a 

voltage-dependent channel is open. During the electrical stimulation, the transmembrane potential 
will also relax approximately exponentially towards a new equilibrium potential. Thus, the 
membrane time constant of the voltage change at the beginning of the stimulus constitutes a 
measurement of the time-averaged membrane resistance. Using appropriate scaling factors to 

30 account for the fraction of the time that the channel is actually open, we can make a quantitative 
estimate of the open-channel membrane resistance. 

c) Measurement of release from inactivation time constant 

Opening an inactivation ion channel requires holding the transmembrane potential below a 
threshold for a time on the order of several milliseconds. This release from inactivation has 
35 important physiological implications. For example, release from inactivation forces a refractory 



-60- 



WO 02/08748 PCT/US01/21652 

period which prevents back-propagation of action potentials, and limits the maximum firing rates 
of neurons. Pharmacological manipulation of this property may be therapeutically relevant 

Using repetitive electrical stimulation, we can estimate the average release from 
inactivation time. This can be done by using electric field pulses of variable width. When the 
5 pulse width falls below the release from inactivation time, fewer channels will be activated and the 
transmembrane potential rise in response to the stimulation will drop. 

d) Measurement of the open channel time 

The open channel time x 0 p en is a function of the inactivation properties of the channel. 
We can detect pharmacological manipulation of this parameter in a medium- to high-throughput 

10 mode by stimulating at very high frequency. For example, consider an assay for a voltage- 
dependent sodium channel using the multiple stimulus method. With a fixed monophasic square 
wave stimulus kernel repeated at a steady rate, the voltage response increases as the stimulus 
repetition rate increases. This is because the sodium channel spends relatively more time open at 
higher frequency. However, if the inter-pulse interval becomes shorter than the open channel time, 

15 the activated sodium channels will be driven negative, and thereby deactivated, by the subsequent 
stimulus pulse. The stimulation burst frequency at which the response flattens is related to the open 
channel time. 

e) Electrical stimulation as an extracellular current damn device 

In whole-cell recording, current clamp is a mode in which command currents can be driven 

20 into the cell while recording the transmembrane potential. Although patch-clamp recording is 
extremely precise, it is a very low-throughput technique. At an absolute maximum under perfect 
conditions, a highly trained scientist could determine cellular parameters at a rate of about ten cells 
per hour. Often, the level of detail obtained with the patch-clamp technique is not necessary for 
drug screening, but there is currently no method for exchanging detail for speed. High speed is 

25 absolutely crucial for screening large compound libraries. 

The electric field stimulation techniques discussed herein permit a new type of current- 
clamp electrophysiology which we call extracellular current clamp. Voltage-dependent channels 
can be used to drive command currents into cell cultures, allowing determination of several cellular 
and channel properties. Extracellular current clamp has a very high throughput, so that it will be 

30 possible to obtain high information content of the pharmacological effects of compound libraries 
against specific ion channel targets. The pharmacology and physiology of a channel can be studied 
directly, or the channel can be used as a current generator for the study of the cell membrane itself 
or a second ion channel. 

While the ultimate precision of the microscopic parameters obtainable with the 

35 extracellular current clamp cannot yet approach the patch-clamp method, we now have the ability 
to exchange information content for throughput. That is, the degree of precision at which to make 



-61- 



WO 02/08748 PCT/US01/21652 

measurements can be arbitrarily set. With a single set of stimulus parameters, large libraries can 
be screened for potential interesting compounds. A medium throughput secondary screen using a 
titration of compound concentrations can be performed on the hits to determine potency and 
specificity. Finally, we can determine such therapeutically relevant properties such as use- 
5 dependence and mechanism of action by varying the stimulus parameters in the presence of the 
compounds. At every stage, the measurements are automatically averaged over many cells, greatly 
reducing uncertainties associated with cell-to-cell variability. 

There are at least two additional advantages of the extracellular current clamp as compared 
to patch-clamp analysis. First, the integrity of the cell membrane is not altered during electric field 

10 stimulation. The intracellular fluid is completely replaced with pipette solution during whole-cell 
patch clamp recording. Many proteins within the cell, including ion channels, are extremely 
sensitive to modulators, intracellular messengers, and the ionic environment. The components of 
the cytoplasm are only crudely known, so the soluble components in the intracellular space are 
always altered. Therefore, the 'normal' physiological state of the cell is only approximated during 

15 whole-cell patch clamp analysis, but remains intact when using extracellular current clamp. 

Second, most cells experience dramatic alterations in gene expression and behavior when 
in contact with other cells. Because most cells also make gap junctional connections with 
neighboring cells, whole-cell patch clamp analysis is only reliable when cells are completely 
isolated from each other. Extracellular current clamp can be used on cells independently of their 

20 degree of confluence, so the cells may be more physiologically relevant. We can use extracellular 
current clamp to find out if there are any effects of cell-cell contact on channel electrophysiology. 
Then, in conjunction with gene expression analysis, we can relate these changes to regulatory 
components of the cell. 

f) Electrical stimulation as an extracellular voltage clamp device 

25 In voltage-clamp, the transmembrane potential of the cell is controlled while monitoring 

the current flow. Voltage clamp is generally achieved by adding a feedback loop to a current 
clamp circuit. In the case of the whole-cell method, this can easily be achieved with the use of two 
pipettes simultaneously attached to the same cell. One pipette passes a command current, while 
the other senses the voltage. A feedback circuit compares the measured voltage with the command 

30 voltage, and adjusts the command current accordingly. Generally, because the cell membrane 
resistance is large compared to the access resistance of the pipette, the same pipette can be used to 
command the current and measure the voltage. Compared to current clamp, voltage clamp is 
generally a more powerful method for electrophysiological analysis. Ion channels are extremely 
sensitive to transmembrane potential, so that analysis of data is far more straightforward when 

35 dealing with current measurements at a fixed voltage. 



-62- 



WO 02/08748 PCT/US01/21652 

Extracellular current clamp can be converted to a voltage clamp method by adding a 
feedback loop between the voltage measurement (the fluorescence of the sensor dye) and the 
current generator (the stimulus parameters). In this case, a transmembrane potential dye with 
sufficient speed is required. The dye combination CC2-DMPE/DiSBAC6(3) has a submillisecond 
5 time constant and should be sufficiently fast to capture all but the fastest cellular events. Based 
upon the difference of the command voltage and the transmembrane potential measurements, a 
computer will alter the stimulus parameters. The stimulus parameters are related to the current 
driven into the cell, so we can determine the time course of the current as a function of the 
command voltage. This method should prove useful in determining the mechanism of action of 

10 pharmacological agents upon ion channels targets. 

g) Assays for intracellular compartments 

The stimulation methods described herein can also be used to modulate the transmembrane 
potentials of intracellular organelles that have phospholipid membranes, including the 
mitochondria and the nucleus. This can be accomplished by first increasing the conductance of the 

15 plasma membrane either by electropermeablization or through the addition of ionophores such as 
valinomycin or gramicidin A. Then, the intracellular space is no longer insulated from the applied 
electric field. This allows an electric field applied to the saline to generate transmembrane 
potential changes across the membranes of intracellular organelles. Then, by staining the cells 
with dyes which are sensitive to the ion concentration or transmembrane potential, and which are 

20 targeted only to the specific organelle membrane of interest, the methods presented herein can be 
used to modulate and assay the ion channels of these organelles. Targeting can be achieved, for 
example via the use of a naturally fluorescent protein containing suitable subcellular location 
signals as are known in the art. 
IX. Introduction of exogenous molecules 

25 Dielectric breakdown of mammalian cell membranes occurs if the electric potential across 

the membrane exceeds about 200 mV (Teissie and Rols, 1993, Biophys. J. 65:409-413). When the 
membrane breaks down, pores are formed through the membrane, bridging the intracellular and 
extracellular spaces. The number and size of the pores increases with increasing transmembrane 
potentials (Kinoshita and Tsong, 1977, Nature 268:438-441). Increasing the electric field strength 

30 above about 60 V/cm on typical mammalian cell lines can electropermeablize the cells. At 
relatively low fields, small pores are created in the cell membrane which apparently are large 
enough to admit small ions, but not large enough to admit molecules as large as DNA (Tsong, 
1991, Biophys. J. 60:297-306). These pores totally depolarize the cell, driving the transmembrane 
potential to near zero. By electropermeablizing cells and monitoring the transmembrane potential 

35 change with a voltage- sensitive dye, the present invention can be used to determine the resting 
transmembrane potential of a cell. This will be useful for determining pharmacological 



-63- 



WO 02/08748 PCT/US01/21652 

interactions with cells or ion channels, either as a primary or a secondary screen. For example, in a 
compound screen against a voltage-dependent sodium channel, one could perform a multiple 
stimulus protocol to determine channel activity. Then, by following with a permeablizing 
protocol, one could determine whether or not the cell membrane had a normal resting potential in 
5 the presence of the compound. 

Additionally, using a highly polarized cell line such as RBL cells, voltage sensitive dyes 
could be easily calibrated by electropermeablization. The starting transmembrane potential under 
various conditions (for example, various concentrations of extracellular potassium), and the final 
transmembrane potential after electropermeablization is zero. 

10 Additionally, the size of the pores created by electropermeablization increases as a 

function of the applied electric field. Below 50 V/cm, no pores are created. Between about 60 
V/cm and 100 V/cm, pores large enough to admit monovalent ions are created. Above around 600 
V/cm, pores large enough to admit DNA are created (Tsong, 1991, Biophys. J. 60:297-306). 
Thus, this invention can be used to create pores of defined size in the cell membranes, in a high- 

15 throughput manner. This could be useful for many applications, including delivery to the 
intracellular space of impermeant ions, impermeant test compounds or other modulators, DNA or 
RNA for the purpose of transient or stable transfection, and fluorescent or other indicator dyes. 
X. Drug Discovery and Screening 
a) Drug Screening 

20 The present invention provides for the reliable detection of test compounds that modulate 

ion channel function that is significantly more versatile and robust than previous assay systems. 
Importantly, the present invention provides the ability to modulate the transmembrane potential in 
intact cells without the requirement of pharmacological agents, or membrane destruction, and loss 
of intracellular contents, as in patching clamping. By providing the ability to externally modulate 

25 the transmembrane potential of living cells, the present invention enables a wide variety of ion 
channels to be assayed. 

Furthermore, this ability to modulate precisely the voltage dependent state of an ion 
channel, has important advantages for drug discovery where it provides the opportunity to screen 
for compounds that interact preferentially with one state, (i.e. use-dependent blockers). For 

30 example, several known therapeutically useful drugs (including anti-arrhythmics, anti-convulsants, 
and local anesthetics) are known to function as use-dependent blockers of voltage-dependent 
sodium and/or calcium channels. In each case, total blockade of the targeted channel would 
typically result in death. Certain conditions, such as chronic pain, arrhythmia, and convulsions 
occur when ceils become over-active. These conditions can be alleviated or eliminated by 

35 blocking the channels if they begin to open too often. Compounds that are capable of blocking the 
channel, but which bind preferentially to the activated or inactivated states(s) rather than the 



-64- 



WO 02/08748 PCT/US01/21652 

resting state(s), can reduce the excitability of muscle and neurons. These drugs are effective 
because they do not affect the channel under normal circumstances, but block it only when 
necessary to prevent hyper-excitability. However existing methods of analysis that are compatible 
with high throughput screening do not provide the ability to routinely control the activation state of 
5 the ion channel in real time. 

In particular, the present invention provides for a method for screening the effect of a test 
compound on an ion channel in a defined functional state within a cell. The method involves 
modulating the transmembrane potential of the cell via the use of repetitive electrical stimulation 
to cycle the ion channel of interest through its activation cycle and to set the transmembrane 
10 potential to a desired level suitable for a specific activation state, or transition between states. 
Then, during or after this process a test compound is added to the cell, and the transmembrane 
potential is measured. 

Typically the results obtained in the presence of the test compound will be compared to a 
control sample incubated in the absence of the test compound. Control measurements are usually 

15 performed with a sample containing all components and under the same stimulation conditions, as 
for the test sample except for the putative drug. Additional control studies can be carried out with 
the ion channel in another voltage dependent state to specifically identify state specific test 
compounds. Detection of a change in transmembrane potential in the presence of the test agent 
relative to the control indicates that the test agent is active and specific on the ion channel in that 

20 state, or during the transition from one state to another. 

Transmembrane potentials can be also be determined in the presence or absence of a 
pharmacologic agent of known activity (i.e., a standard agent) or putative activity (i.e., a test 
agent). A difference in transmembrane potentials as detected by the methods disclosed herein 
allows one to compare the activity of the test agent to that of the standard agent. It will be 

25 recognized that many combinations and permutations of drug screening protocols are known to one 
of skill in the art and they may be readily adapted to use with the present inventions disclosed 
herein to identify compounds, which affect ion channels and or transmembrane potentials. Use of 
the present inventions in combination with all such methods are contemplated by this invention. 

In another aspect the present invention includes the use of a second ion channel in 

30 conjunction with electrical stimulation methods described herein to set the resting, or stimulated 
transmembrane potential to a predefined value thereby providing for the ability to assay a first ion 
channel of interest. In one embodiment the second ion channel is a voltage regulated sodium or 
calcium channel which enables the generation of sustained positive transmembrane potentials. In 
another embodiment the second ion channel is a voltage regulated potassium channel, enabling the 

35 generation of negative transmembrane potentials. The use of these second ion channels enables the 



-65- 



WO 02/08748 PCT/US01/21652 

electrical stimulation method to be used to set the transmembrane potential to virtually any 
predefined level. 

Because this assay format involves two ion channels, modulators of either channel will 
affect the voltage response. In this case additional control studies may be carried out with the 
5 parental cell line expressing only the second ion channel used to set the transmembrane potential. 
Compounds that block the first ion channel can then be re-tested separately to find out if they have 
activity against the second ion channel. 

Typically the test compounds screened will be present in libraries of related or diverse 
compounds. The library can have individual members that are tested individually or in 
10 combination, or the library can be a combination of individual members. Such libraries can have 
at least two members, preferably greater than about 100 members or greater than about 1,000 
members, more preferably greater than about 1 0,000 members, and most preferably greater than 
about 100,000 or 1,000,000 members. 

b) Selectivity and Toxicology of Candidate Modulators 

15 Once identified, candidate modulators can be evaluated for selectivity and toxicological 

effects using known methods (see, Lu, Basic Toxicology, Fundamentals, Target Organs, and Risk 
Assessment, Hemisphere Publishing Corp., Washington (1985); U.S. Patent Nos: 5,196,313 to 
Culbreth (issued March 23, 1993) and U.S. Patent No. 5,567,952 to Benet (issued October 22, 
1996). 

20 For example primary cell lines, or tissue slices can be used to screen for the effect of the 

candidate modulator on the response of the ion channel of interest in its native physiological 
context. For example, to screen for drugs that exhibit specific, and/or selective effects on heart 
cells it may be preferable to use myocytes or other in vitro cell culture model cell lines. In this 
case, a primary screen could be completed in a myocyte derived cell line to identify compounds 

25 that either shorten, prolong or block electrically-induced action potentials. 

The secondary screen would then be designed to identify compounds that exhibit 
potentially adverse effects on the body. For example, this can be accomplished by screening for 
the effects of the candidate drug on electrically excitable tissues such as heart or neuronal tissues, 
or immortalized cell cultures derived from these tissues. These tissues play critical roles within an 

30 organism and any undesired effect of the candidate drug on the ability of these tissues to be 
electrically stimulated would be predicted to create potential serious side effects when 
administered. As a consequence, active compounds that also impaired the ability of these tissues 
to function could be eliminated from consideration as a drug candidate at an early stage, or have 
medicinal chemistry performed to reduce the side effects. 

35 Additional toxicological analysis of candidate modulators can be established by 

determining in vitro toxicity towards a cell line, such as a mammalian (preferably human) cell 



-66- 



WO 02/08748 PCT/US01/21652 

line. Candidate modulators can be treated with, for example, tissue extracts, such as preparations of 
liver, such as microsomal preparations, to determine increased or decreased toxicological 
properties of the chemical after being metabolized by a whole organism, or via their ability to be 
degraded via Cytochrome P450 systems as described in commonly owned U.S. Patent Application 
5 no. 09/301,525, filed April 28, 1999, U.S. Patent Application No. 09/301,395 filed April 28, 1999 
and U.S. Application No. 09/458,927 filed December 10, 1999. The results of these types of 
studies are often predictive of toxicological properties of chemicals in animals, such as mammals, 
including humans. 

The toxicological activity can be measured using reporter genes that are activated during 
10 toxicological activity or by cell lysis (see WO 98/13353, published 4/2/98). Preferred reporter 
genes produce a fluorescent or luminescent translational product (such as, for example, a Green 
Fluorescent Protein (see, for example, U.S. Patent No. 5,625,048 to Tsien et al, issued 4/29/98; 
U.S. Patent No. 5,777,079 to Tsien et al, issued 7/7/98; WO 96/23810 to Tsien, published 8/8/96; 
WO 97/28261, published 8/7/97; PCT/US97/12410, filed 7/16/97; PCT/US97/14595, filed 
15 8/15/97)) or a translational product that can produce a fluorescent or luminescent product (such as, 
for example, beta- lactamase (see, for example, U.S. Patent No. 5,741,657 to Tsien, issued 4/21/98, 
and WO 96/30540, published 10/3/96)), such as an enzymatic degradation product. Cell lysis can 
be detected in the present invention as a reduction in a fluorescence signal from at least one 
photon-producing agent within a cell in the presence of at least one photon reducing agent. Such 
20 toxicological determinations can be made using prokaryotic or eukaryotic cells, optionally using 
toxicological profiling, such as described in PCT/US94/00583, filed 1/21/94 (WO 94/17208), 
German Patent No 69406772.5-08, issued 11/25/97; EPC 0680517, issued 11/12/94; U.S. Patent 
No. 5,589,337, issued 12/31/96; EPO 651825, issued 1/14/98; and U.S. Patent No. 5,585,232, 
issued 12/17/96). 

25 Alternatively, or in addition to these in vitro studies, the bioavailability and toxicological 

properties of a candidate modulator in an animal model, such as mice, rats, rabbits, or monkeys, 
can be determined using established methods (see, Lu, supra (1985); and Creasey, Drug 
Disposition in Humans, The Basis of Clinical Pharmacology, Oxford University Press, Oxford 
(1979), Osweiler, Toxicology , Williams and Wilkins, Baltimore, MD (1995), Yang, Toxicology of 

30 Chemical Mixtures; Case Studies, Mechanisms, and Novel Approaches, Academic Press, Inc., San 
Diego, CA (1994), Burrell et ah, Toxicology of the Immune System; A Human Approach, Van 
Nostrand Reinhld, Co. (1997), Niesink et al., Toxicology; Principles and Applications, CRC Press, 
Boca Raton, FL (1996)). Depending on the toxicity, target organ, tissue, locus, and presumptive 
mechanism of the candidate modulator, the skilled artisan would not be burdened to determine 

35 appropriate doses, LD50 values, routes of administration, and regimes that would be appropriate to 
determine the toxicological properties of the candidate modulator. In addition to animal models, 



-67- 



WO 02/08748 PCT/US01/21652 

human clinical trials can be performed following established procedures, such as those set forth by 
the United States Food and Drug Administration (USFDA) or equivalents of other governments. 
These toxicity studies provide the basis for determining the therapeutic utility of a candidate 
modulator in vivo. 

5 c) Efficacy of Candidate Modulators 

Efficacy of a candidate modulator can be established using several art-recognized methods, 
such as in vitro methods, animal models, or human clinical trials (see, Creasey, supra (1979)). 
Recognized in vitro models exist for several diseases or conditions. For example, the ability of a 
chemical to extend the life-span of HIV-infected cells in vitro is recognized as an acceptable model 

10 to identify chemicals expected to be efficacious to treat HIV infection or AIDS (see, Daluge et aL 9 
Antimicro. Agents Chemothen 41:1082-1093 (1995)). Furthermore, the ability of cyclosporin A 
(CsA) to prevent proliferation of T-cells in vitro has been established as an acceptable model to 
identify chemicals expected to be efficacious as immunosuppressants (see, Suthanthiran et aL, 
supra, (1996)). For nearly every class of therapeutic, disease, or condition, an acceptable in vitro 

15 or animal model is available. Such models exist, for example, for gastro-intestinal disorders, 
cancers, cardiology, neurobiology, and immunology. In addition, these in vitro methods can use 
tissue extracts, such as preparations of liver, such as microsomal preparations, to provide a reliable 
indication of the effects of metabolism on the candidate modulator. Similarly, acceptable animal 
models may be used to establish efficacy of chemicals to treat various diseases or conditions. For 

20 example, the rabbit knee is an accepted model for testing chemicals for efficacy in treating arthritis 
(see, Shaw and Lacy, J. Bone Joint Surg. (Br) 55:197-205 (1973)). Hydrocortisone, which is 
approved for use in humans to treat arthritis, is efficacious in this model which confirms the 
validity of this model (see, McDonough, Phys. Then 62:835-839 (1982)). When choosing an 
appropriate model to determine efficacy of a candidate modulator, the skilled artisan can be guided 

25 by the state of the art to choose an appropriate model, dose, and route of administration, regime, 
and endpoint and as such would not be unduly burdened. 

In addition to animal models, human clinical trials can be used to determine the efficacy of 
a candidate modulator in humans. The USFDA, or equivalent governmental agencies, have 
established procedures for such studies (see, www.fda.govl 

30 d) Selectivity of Candidate Modulators 

The in vitro and in vivo methods described above also establish the selectivity of a 
candidate modulator. The present invention provides a rapid method of determining the specificity 
of the candidate modulator. For example, cell lines containing related ion channel family members 
can be used to rapidly profile the selectivity of a test chemical with respect both to its ability to 

35 inhibit related ion channels, and their relative ability to modulate different voltage dependent states 
of the ion channels. Such a system provides for the first time the ability to rapidly profile large 



-68- 



WO 02/08748 PCT/US01/21652 

numbers of test chemicals in order to systematically evaluate in a simple, miniaturized high 
throughput format the ion channel selectivity of a candidate modulator. 

e) An identified chemical, modulator, or therapeutic and compositions 

The invention includes compositions, such as novel chemicals, and therapeutics identified 
5 by at least one method of the present invention as having activity by the operation of methods, 
systems or components described herein. Novel chemicals, as used herein, do not include 
chemicals already publicly known in the art as of the filing date of this application. Typically, a 
chemical would be identified as having activity from using the invention and then its structure 
revealed from a proprietary database of chemical structures or determined using analytical 

10 techniques such as mass spectroscopy. 

One embodiment of the invention is a chemical with useful activity, comprising a chemical 
identified by the method described above. Such compositions include small organic molecules, 
nucleic acids, peptides and other molecules readily synthesized by techniques available in the art 
and developed in the future. For example, the following combinatorial compounds are suitable for 

15 screening: peptoids (PCT Publication No. WO 91/19735, 26 Dec. 1991), encoded peptides (PCT 
Publication No. WO 93/20242, 14 Oct. 1993), random bio-oligomers (PCT Publication WO 
92/00091, 9 Jan. 1992), benzodiazepines (U.S. Pat. No. 5,288,514), diversomeres such as 
hydantoins, benzodiazepines and dipeptides (Hobbs DeWitt, S. et ah, Proc. Nat. Acad. Sci. USA 
90: 6909-6913 (1993)), vinylogous polypeptides (Hagihara et aL, J. Amer. Chem. Soc. 114: 6568 

20 (1992)), nonpeptidal peptidomimetics with a Beta-D-Glucose scaffolding (Hirschmann, R. et al., J. 
Amer. Chem. Soc. 114: 9217-9218 (1992)), analogous organic syntheses of small compound 
libraries (Chen, C. et ah, J. Amer. Chem. Soc. 116:2661 (1994)), oligocarbamates (Cho, C.Y. et 
al., Science 261: 1303 (1993)), and/or peptidyl phosphonates (Campbell, D.A. et ah, J. Org. Chem. 
59: 658 (1994)). See, generally, Gordon, E. M. et ah, J. Med Chem. 37: 1385 (1994). 

25 The present invention also encompasses the identified compositions in a pharmaceutical 

composition comprising a pharmaceutically acceptable carrier prepared for storage and subsequent 
administration, which have a pharmaceutically effective amount of the products disclosed above in 
a pharmaceutically acceptable carrier or diluent. Acceptable carriers or diluents for therapeutic use 
are well known in the pharmaceutical art, and are described, for example, in Remington's 

30 Pharmaceutical Sciences, Mack Publishing Co. (A.R. Gennaro edit. 1985). Preservatives, 
stabilizers, dyes and even flavoring agents may be provided in the pharmaceutical composition. 
For example, sodium benzoate, acsorbic acid and esters of p-hydroxybenzoic acid may be added as 
preservatives. In addition, antioxidants and suspending agents may be used. 

The compositions of the present invention may be formulated and used as tablets, capsules 

35 or elixirs for oral administration; suppositories for rectal administration; sterile solutions, 
suspensions for injectable administration; and the like. Injectables can be prepared in conventional 



-69- 



WO 02/08748 PCT/US01/21652 

forms, either as liquid solutions or suspensions, solid forms suitable for solution or suspension in 
liquid prior to injection, or as emulsions. Suitable excipients are, for example, water, saline, 
dextrose, mannitol, lactose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, and the 
like. In addition, if desired, the injectable pharmaceutical compositions may contain minor 
5 amounts of nontoxic auxiliary substances, such as wetting agents, pH buffering agents, and the 
like. If desired, absorption enhancing preparations (e.g., liposomes), may be utilized. 

The pharmaceutically effective amount of the composition required as a dose will depend 
on the route of administration, the type of animal being treated, and the physical characteristics of 
the specific animal under consideration. The dose can be tailored to achieve a desired effect, but 

10 will depend on such factors as weight, diet, concurrent medication and other factors which those 
skilled in the medical arts will recognize. In practicing the methods of the invention, the products 
or compositions can be used alone or in combination with one another, or in combination with 
other therapeutic or diagnostic agents. These products can be utilized in vivo, ordinarily in a 
mammal, preferably in a human, or in vitro. In employing them in vivo, the products or 

15 compositions can be administered to the mammal in a variety of ways, including parenterally, 
intravenously, subcutaneously, intramuscularly, colonically, rectally, nasally or intraperitoneally, 
employing a variety of dosage forms. Such methods may also be applied to testing chemical 
activity in vivo. 

As will be readily apparent to one skilled in the art, the useful in vivo dosage to be 

20 administered and the particular mode of administration will vary depending upon the age, weight 
and mammalian species treated, the particular compounds employed, and the specific use for 
which these compounds are employed. The determination of effective dosage levels, that is the 
dosage levels necessary to achieve the desired result, can be accomplished by one skilled in the art 
using routine pharmacological methods. Typically, human clinical applications of products are 

25 commenced at lower dosage levels, with dosage level being increased until the desired effect is 
achieved. Alternatively, acceptable in vitro studies can be used to establish useful doses and routes 
of administration of the compositions identified by the present methods using established 
pharmacological methods. 

In non-human animal studies, applications of potential products are commenced at higher 

30 dosage levels, with dosage being decreased until the desired effect is no longer achieved or adverse 
side effects disappear. The dosage for the products of the present invention can range broadly 
depending upon the desired affects and the therapeutic indication. Typically, dosages may be 
between about 10 jag/kg and 100 mg/kg body weight, and preferably between about 100 jig/kg and 
10 mg/kg body weight. Administration is preferably oral on a daily basis. 

35 The exact formulation, route of administration and dosage can be chosen by the individual 

physician in view of the patient's condition. (See e.g., Fingl et ah, in The Pharmacological Basis 



-70- 



WO 02/08748 PCT/US01/21652 

of Therapeutics, 1975). It should be noted that the attending physician would know how to and 
when to terminate, interrupt, or adjust administration due to toxicity, or to organ dysfunctions. 
Conversely, the attending physician would also know to adjust treatment to higher levels if the 
clinical response were not adequate (precluding toxicity). The magnitude of an administrated dose 
5 in the management of the disorder of interest will vary with the severity of the condition to be 
treated and to the route of administration. The severity of the condition may, for example, be 
evaluated, in part, by standard prognostic evaluation methods. Further, the dose and perhaps dose 
frequency, will also vary according to the age, body weight, and response of the individual patient. 
A program comparable to that discussed above may be used in veterinary medicine. 

10 Depending on the specific conditions being treated, such agents may be formulated and 

administered systemically or locally. Techniques for formulation and administration may be found 
in Remington's Pharmaceutical Sciences, 18th Ed., Mack Publishing Co., Easton, PA (1990). 
Suitable routes may include oral, rectal, transdermal, vaginal, transmucosal, or intestinal 
administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary 

15 injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or 
intraocular injections. 

For injection, the agents of the invention may be formulated in aqueous solutions, 
preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or 
physiological saline buffer. For such transmucosal administration, penetrants appropriate to the 

20 barrier to be permeated are used in the formulation. Such penetrants are generally known in the 
art. Use of pharmaceutically acceptable carriers to formulate the compounds herein disclosed for 
the practice of the invention into dosages suitable for systemic administration is within the scope 
of the invention. With proper choice of carrier and suitable manufacturing practice, the 
compositions of the present invention, in particular, those formulated as solutions, may be 

25 administered parenterally, such as by intravenous injection. The compounds can be formulated 
readily using pharmaceutically acceptable carriers well known in the art into dosages suitable for 
oral administration. Such carriers enable the compounds of the invention to be formulated as 
tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by 
a patient to be treated. 

30 Agents intended to be administered intracellularly may be administered using techniques 

well known to those of ordinary skill in the art. For example, such agents may be encapsulated 
into liposomes, then administered as described above. All molecules present in an aqueous 
solution at the time of liposome formation are incorporated into the aqueous interior. The 
liposomal contents are both protected from the external micro-environment and, because liposomes 

35 fuse with cell membranes, are efficiently delivered into the cell cytoplasm. Additionally, due to 
their hydrophobicity, small organic molecules may be directly administered intracellularly. 



-71- 



WO 02/08748 PCT/US01/21652 

Pharmaceutical compositions suitable for use in the present invention include compositions 
wherein the active ingredients are contained in an effective amount to achieve its intended purpose. 
Determination of the effective amounts is well within the capability of those skilled in the art, 
especially in light of the detailed disclosure provided herein. In addition to the active ingredients, 
5 these pharmaceutical compositions may contain suitable pharmaceutically acceptable carriers 
comprising excipients and auxiliaries which facilitate processing of the active compounds into 
preparations which can be used pharmaceutically. The preparations formulated for oral 
administration may be in the form of tablets, dragees, capsules, or solutions. The pharmaceutical 
compositions of the present invention may be manufactured in a manner that is itself known, for 

10 example, by means of conventional mixing, dissolving, granulating, dragee-making, levitating, 
emulsifying, encapsulating, entrapping, or lyophilizing processes. 

Pharmaceutical formulations for parenteral administration include aqueous solutions of the 
active compounds in water-soluble form. Additionally, suspensions of the active compounds may 
be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles 

15 include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or 
triglycerides, or liposomes. Aqueous injection suspensions may contain substances that increase 
the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. 
Optionally, the suspension may also contain suitable stabilizers or agents that increase the 
solubility of the compounds to allow for the preparation of highly concentrated solutions. 

20 Pharmaceutical preparations for oral use can be obtained by combining the active 

compounds with solid excipient, optionally grinding a resulting mixture, and processing the 
mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. 
Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or 
sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato 

25 starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium 
carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may 
be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such 
as sodium alginate. Dragee cores are provided with suitable coatings. For this purpose, 
concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, 

30 polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer 
solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added 
to the tablets or dragee coatings for identification or to characterize different combinations of 
active compound doses. For this purpose, concentrated sugar solutions may be used, which may 
optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, 

35 and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. 
Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to 



-72- 



WO 02/08748 PCT/US01/21652 

characterize different combinations of active compound doses. Such formulations can be made 
using methods known in the art (see, for example, U.S. Patent Nos. 5,733,888 (injectable 
compositions); 5,726,181 (poorly water soluble compounds); 5,707,641 (therapeutically active 
proteins or peptides); 5,667,809 (lipophilic agents); 5,576,012 (solubilizing polymeric agents); 
5 5,707,615 (anti-viral formulations); 5,683,676 (particulate medicaments); 5,654,286 (topical 
formulations); 5,688,529 (oral suspensions); 5,445,829 (extended release formulations); 5,653,987 
(liquid formulations); 5,641,515 (controlled release formulations) and 5,601,845 (spheroid 
formulations). 

XI. Embodiments of the Invention 
10 Some of the embodiments of the present invention are as follows: 

A method of characterizing the biological activity of a candidate compound comprising: 
exposing one or more cells to said compound; 

repetitively exposing said one or more cells to one or more electric fields so as to effect a 
controlled change in transmembrane potential of said one or more cells; and 
1 5 monitoring, without using a patch clamp, changes in the transmembrane potential of said 

one or more cells. 

The above method, where the monitoring comprises detecting fluorescence emission from 
an area of observation containing said one or more cells. 

The above method, where the electric fields are biphasic. 
20 The above method, additionally comprising limiting spatial variation in electric field 

intensity so as to minimize irreversible cell electroporation. 

The above method, where one or more electrical fields cause an ion channel of interest to 
cycle between different voltage dependent states. 

The above method, where the one or more electrical fields cause an ion channel of interest 

25 to open. 

The above method, where the one or more electrical fields cause an ion channel of interest 
to be released from inactivation. 

The above method, where the one or more cells comprise a voltage sensor selected from 
the group consisting of a FRET based voltage sensor, an electrochromic transmembrane potential 
30 dye, a transmembrane potential redistribution dye, an ion sensitive fluorescent or luminescent 
molecule and a radioactive ion. 

The above method, where the one or more cells comprise a voltage regulated ion channel. 
The above method, where the voltage regulated ion channel is selected from the group 
consisting of a potassium channel, a calcium channel, a chloride channel and a sodium channel, 
35 The above method, where the electric field exhibits limited spatial variation in intensity in 

the area of observation of less than about 25% from a mean intensity in that area. 



-73- 



WO 02/08748 PCT/US01/21652 

The above method, where the one or more electrical fields varies over an area of 
observation by no more than about 15 % from the mean electrical field at any one time. 

The above method, where the one or more electrical fields varies over an area of 
observation by no more than about 5 % from the mean electrical field at any one time. 
5 The above method, where the one or more electrical fields comprises stimulation with 

either a square wave-form, a sinusoidal wave-form or a saw tooth wave-form. 

The above method, where the one or more electrical fields have an amplitude within the 
range of about 10 V/cm to about 100 V/cm, 

The above method, where the one or more electrical fields have an amplitude within the 
10 range of about 20 V/cm to about 80 V/cm. 

The above method, where the one or more electrical fields are repeated at a frequency of 
stimulation that is greater than or equal to the reciprocal of the transmembrane time constant of 
said one or more cells. 

The above method, where the one or more electrical fields are repeated at a frequency of 
1 5 stimulation within the range of zero to 1kHz. 

The above method, where the one or more electrical fields have a pulse duration within the 
range of about 100 microseconds to about 20 milliseconds. 

The above method, where the transmembrane potential is developed across the plasma 
membrane of said one or more cells. 
20 A method of assaying the biochemical activity of a compound against a target ion channel 

comprising: 

selecting a cell line having a normal resting transmembrane potential corresponding to a 
selected voltage dependent state of said target ion channel; 

expressing said target ion channel in a population of cells of said selected cell line; 
25 exposing said population of cells to said compound; 

repetitively exposing said population of cells to one or more electric fields so as to effect a 
controlled change in transmembrane potential of said one or more cells; and 

monitoring changes in the transmembrane potential of said one or more cells. 

The above method, where the target ion channel is exogenously expressed in said cell line. 
30 The above method, where the cell line is transfected with nucleic acid encoding said target 

ion channel. 

The above method, where the cell line expresses insignificant levels of other ion channels. 
The above method, where the cell line is selected from the group consisting of CHL, 
LTK(-),and CHO-K1. 



-74- 



WO 02/08748 PCT/US01/21652 

The above method, where the target ion channel is a sodium channel, and wherein said 
population of cells is selected from the group consisting of CHL cells, LTK(-) cells, and CHO-K1 
cells. 

The above method, where the target ion channel is a sodium channel, and wherein said 
5 population of cells is selected from the group consisting of HEK-293 cells, RBL cells, Fll cells, 
and HL5 cells. 

The above method, where the target ion channel is a potassium channel, and wherein said 
population of cells is selected from the group consisting of CHL cells, LTK(-) cells, and CHO-K1 
cells. 

10 The above method, where the target ion channel is a calcium channel, and wherein said 

population of cells is selected from the group consisting of CHL cells, LTK(-) cells, and CHO-K1 
cells. 

A method of assaying ion channel activity comprising: 

exposing at least one cell to a plurality of electric field pulses so as to create a controlled 
1 5 change in transmembrane potential and so as to activate an ion channel of interest; and 

detecting ion channel activity by detecting one or more changes in transmembrane 
potential without using a patch clamp. 

The above method, where the at least one cell comprises a voltage sensor selected from the 
group consisting of a FRET based voltage sensor, an electrochromic transmembrane potential dye, 
20 a transmembrane potential redistribution dye, an ion sensitive fluorescent or luminescent molecule 
and a radioactive ion. 

The above method, where the voltage sensor comprises a FRET based voltage sensor. 

The above method, where the ion channel of interest is a voltage regulated ion channel. 

The above method, where the plurality of electric field pulses cause said ion channel of 
25 interest to cycle between different voltage dependent states. 

The above method, where the at least one cell is an eukaryotic cell. 

The above method, where the at least one cell is a non-excitable cell. 

The above method, where the at least one cell is a prokaryotic cell. 

The above method, where the at least one cell is a tissue culture cell. 
30 The above method, where the at least one cell is a primary cell line. 

The above method, where the at least one cell is part of an intact living organism. 

A method of assaying ion channel activity comprising: 

expressing a selected target ion channel in at least one cell; 

expressing a selected counter ion channel in said at least one cell; 
35 exposing said at least one cell to a plurality of electric field pulses so as to create a 

controlled change in transmembrane potential and so as to activate said counter ion channel; and 



-75- 



WO 02/08748 PCT/US01/21652 
monitoring the transmembrane potential of said at least one celL 

The above method, where a transmembrane potential change is detected when said ion 
channel of interest is blocked. 

The above method, where the ion channel of interest comprises a ligand gated ion channel. 
5 The above method, where the counter channel comprises a sodium channel. 

A method of modifying the transmembrane potential of a cell comprising repetitively 
applying biphasic electric field pulses to said cell, wherein said pulses have a maximum amplitude 
of less than approximately 90 V/cm, wherein said pulses are applied at a rate of at least about 1 per 
second, and wherein the total duration of each pulse is at least about 1 millisecond. 
10 The above method, where the maximum amplitude is approximately 20 to 40 V/cm. 

The above method, where the pulse duration is approximately 2 to 10 milliseconds per 

phase. 

The above method, where the pulses are applied at a rate of approximately 20 to 100 pulses 
per second. 

15 A method of characterizing the biological activity of a candidate compound comprising: 

placing one or more cells into an area of observation in a sample well; 
exposing said one or more cells to said compound; 

repetitively exposing said one or more cells to a series of biphasic electric fields at a rate of 
approximately 20 to 100 pulses per second, wherein said electric fields exhibit limited spatial 
20 variation in intensity in the area of observation of less than about 25% from a mean intensity in 
that area, and wherein said electric fields produce a controlled change in transmembrane potential 
of said one or more cells; and 

monitoring changes in the transmembrane potential of said one or more cells by detecting 
fluorescence emission of a FRET based voltage sensor from an area of observation containing said 
25 one or more cells. 

The above method, where the one or more electrical fields cause an ion channel of interest 

to open. 

The above method, where the one or more electrical fields cause an ion channel of interest 
to be released from inactivation. 
30 The above method, where the one or more cells comprise a voltage regulated ion channel. 

The above method, where the voltage regulated ion channel is selected from the group 
consisting of a potassium channel, a calcium channel, a chloride channel and a sodium channel. 

The above method, where the one or more electrical fields varies over an area of 
observation by no more than about 1 5 % from the mean electrical field at any one time. 
35 The above method, where the one or more electrical fields varies over an area of 

observation by no more than about 5 % from the mean electrical field at any one time. 



-76- 



WO 02/08748 PCT/US01/21652 

The above method, where the one or more electrical fields are selected from a square 
wave-form, a sinusoidal wave-form or a saw tooth wave-form. 
A high throughput screening system comprising: 

a plurality of wells having a high transmittance portion through which cells present in said 
5 wells are optically observable in an area of observation; 

two electrodes in each of said plurality of wells; 

an optical detector configured to detect light emanating from said wells through said high 
transmittance portion; 

a power supply connected to said electrodes; wherein said power supply and said 
10 electrodes are configured to apply a series of electric fields to cells within said area of observation, 
said electric fields having a spatial variation of less than about 25% of a mean field intensity within 
said area of observation, said electric fields being effective to controllably alter the transmembrane 
potential of a portion of said cells; 

a data processing unit configured to interpret said light emanating from said wells through 
15 said high transmittance portion as ion channel activity resulting from said transmembrane potential 
alterations. 

The above high throughput screening system, where the pluarality of wells are located in a 
multiwell plate. 

The above high throughput screening system, where the high transmittance portion is made 
20 from a material selected from the group consisting of glass, quartz, cycloolefln, Aclar, 
polypropylene, polyethylene and polystyrene. 

The above high throughput screening system, where the high transmittance portion 
exhibits less fluorescence when excited with UV light in the range of 250 nm to 400 nm than 
polystyrene. 

25 The above high throughput screening system, where the electrodes are located in a wall of 

said plurality of wells. 

The above high throughput screening system, where the electrodes are located in a bottom 
layer of said plurality of wells. 

The above high throughput screening system, where the multiwell plate comprises up to 96 

30 wells. 

The above high throughput screening system, where the multiwell plate comprises greater 
than 96 wells. 

The above high throughput screening system, where the multiwell plate comprises greater 
than 384 wells. 



-77- 



WO 02/08748 PCT/US01/21652 

The above high throughput screening system, where the electrodes are made of a material 
selected from the group consisting of gold, platinum, palladium, chromium, molybdenum, iridium, 
tungsten, tantalum and titanium. 

The above high throughput screening system, where the multiwell plate comprises 
5 optically opaque materials or pigments to reduce the transmission of light. 

The above high throughput screening system, where the electrodes are separated by a gap 
within the range of about 1 to 4 mm. 

The above high throughput screening system, where the electrodes are separated by a gap 
within the range of about 0.1 to 1 mm. 
10 The above high throughput screening system, where the electrodes are separated by a gap 

within the range of about 0.01 to 0.1 mm. 

The above high throughput screening system, where the electrodes are charged to create an 
electrical field intensity of between 5 to 100 V/cm across said gap, and wherein the total charge 
transferred across the surface area of the electrically conductive material, in fluidic connection 
15 with the interior of the well is less than or equal to lOOjuC/mm 2 . 

The above high throughput screening system, where the plurality of wells further comprise 
an insulator orientated and configured so as to create an area of observation within said well in 
which the electrical field intensity varies by no more than 10 % from the mean electrical field 
intensity when said at least two strips of electrically conductive material are charged to create an 
20 electrical field intensity of between 5 to 100 V/cm across said gap, and wherein the total charge 
transferred across the surface area of the electrically conductive material, in fluidic connection 
with the interior of the well is less than or equal to lOOjuC/mm^ . 

The above high throughput screening system, where the plurality of wells further comprise 
at least two satellite electrical conductors. 
25 A high throughput screening system comprising: 

sample wells; 

liquid handling stations for adding reagents and/or cells to said sample wells; and 
means for controlling the transmembrane potential of cells in said sample wells so as to 
selectively cause ion channel activity. 
30 means for optically monitoring changes in said transmembrane potential. 

The above high throughput screening system, where the means comprises electrodes 
configured to create an electric field having a spatial variation of less than about 25% of a mean 
field intensity within an area of observation. 

The above high throughput screening system, where the means for controlling the 
35 transmembrane potential comprise an electrode array assembly. 



-78- 



WO 02/08748 PCT/US01/21652 

The above high throughput screening system, where the electrode assembly array 
comprises 8 electrode assemblies. 

The above high throughput screening system, where the electrode assembly array 
comprises 96 electrode assemblies. 
5 The above high throughput screening system, where the electrode assembly array 

comprises greater than 96 electrode asemblies. 

The above high throughput screening system, where the system further comprises means 
for retractably moving said electrode assembly into and out of the wells of a multiwell plate. 

The above high throughput screening system, where the means for controlling the 
10 transmembrane potential comprises electrical conductors with two substantially parallel planar 
surfaces. 

The above high throughput screening system, where the electrical conductors are separated 
by a gap within the range of 1 to 4 mm. 

The above high throughput screening system, where the electrical conductors are separated 
15 by a gap within the range of 0.1 to 1 mm. 

The above high throughput screening system, where the electrical conductors further 
comprise a first insulator. 

The above high throughput screening system, where the first insulator comprises two 
planar surfaces orientated perpendicular to said substantially parallel planar surfaces of said 
20 electrical conductors and substantially parallel with respect to each other. 

The above high throughput screening system, where the electrical conductors further 
comprise a second insulator attached to said at least two electrical conductors, wherein said second 
insulator is interposed in said gap between said at least two electrical conductors to define the 
depth of said aqueous solution between said at least two electrical conductors. 
25 The above high throughput screening system, where the first insulator is composed of a 

low fluorescence material, wherein said low fluorescence material exhibits less fluorescence when 
excited with UV light in the range 250 nm to 400 nm than polystyrene of comparable size. 

The above high throughput screening system, where the second insulator is composed of a 
low fluorescence material, wherein said low fluorescence material exhibits less fluorescence when 
30 excited with UV light in the range 250 nm to 400 nm than polystyrene of comparable size. 

The above high throughput screening system, where the first insulator comprises an 
insulator selected from the group consisting of plastic, glass and ceramic. 

The above high throughput screening system, where the plastic is selected from the group 
consisting of nylon, polystyrene, Teflon (tetrafluoroethylene), polypropylene, polyethylene, 
35 polyvinyl chloride, and cycloolefin. 



-79- 



WO 02/08748 PCT/US01/21652 

. The above high throughput screening system, where the electrical conductors comprise a 
conductor selected from the group consisting of gold, platinum, titanium, tungsten, molybdenum, 
iridium, vandium, Nb, Ta, stainless steel and graphite. 

The above high throughput screening system, where the electrical conductors comprise a 
5 surface treatment to reduce electrolysis. 

The above high throughput screening system, where the surface treatment to reduce 
electrolysis comprises platinum black, gold black, iridium/iridium oxide, titanium/titanium nitride 
or polypyrrole films. 

The above high throughput screening system, where the electrical field intensity varies by 
10 no more than 10 % from the mean electrical field intensity when said at least two electrical 
conductors are charged to create an electrical field intensity of between 5 to 100 V/cm across said 
gap, wherein the total charge transferred across the surface area of the electrical conductors in 
contact with said aqueous solution is less than or equal to 100 DC/mm2. 

The above high throughput screening system, where the electrical field intensity varies by 
15 no more than 5% from the mean electrical field intensity when said at least two electrical 
conductors are charged to create an electrical field intensity of between 5 to 100 V/cm across said 
gap, wherein the total charge transferred across the surface area of the electrical conductors in 
contact with said aqueous solution is less than or equal to 100 jnC/mm 2 . 

A method of screening a plurality of drug candidate compounds against a target ion 
20 channel comprising: 

expressing said target ion channel in a population of host cells; 
placing a plurality of said host cells into each of a plurality of sample wells; 
adding a candidate drug compound to at least one of said plurality of sample wells; and 
modulating the transmembrane potential of host cells in said plurality of sample wells with 
25 a repetitive application of electric fields so as to set said transmembrane potential to a level 
corresponding to a pre-selected voltage dependent state of said target ion channel. 

The above method, additionally comprising selecting a host cell line having a normal 
resting transmembrane potential corresponding to a second pre-selected voltage dependent state of 
said target ion channel. 
30 The above method, where the electric fields are biphasic. 

The above method, where electric fields cause an ion channel of interest to cycle between 
different voltage dependent states. 

The above method, where the electric fields cause an ion channel of interest to open. 
The above method, where the electric fields cause an ion channel of interest to be released 
35 from inactivation. 



-80- 



WO 02/08748 PCT/US01/21652 

The above method, where the one or more cells comprise a voltage sensor selected from 
the group consisting of a FRET based voltage sensor, an electrochromic transmembrane potential 
dye, a transmembrane potential redistribution dye, an ion sensitive fluorescent or luminescent 
molecule and a radioactive ion. 
5 The above method, where the target ion channel is selected from the group consisting of a 

potassium channel, a calcium channel, a chloride channel and a sodium channel. 

The above method, where the one or more electrical fields comprises stimulation with 
either a square wave-form, a sinusoidal wave-form or a saw tooth wave-form. 

The above method, where the one or more electrical fields have an amplitude within the 
10 range of about 10 V/cm to about 100 V/cm. 

The above method, where the one or more electrical fields have an amplitude within the 
range of about 20 V/cm to about 80 V/cm. 

An assay plate and electrode assembly comprising at least one sample well having 
electrodes placed therein, wherein said electrodes are positioned with respect to the bottom surface 
15 of the well to provide an electric field adjacent to said bottom surface that varies by less than about 
10% from a mean field intensity over at least about 20% of the surface area of said bottom surface. 

The above assembly, where the electrodes comprise plate electrodes extending down into 
said well such that bottom ends of said electrodes are adjacent to but not in contact with said 
bottom surface. 

20 The above assembly, comprising two electrodes per sample well. 

The above assembly, comprising more than two electrodes per sample well. 

The above assembly, where the electrodes are plated onto said bottom surface of said well. 

The above assembly, where the bottom surface comprises a high optical transmittance 

portion. 

25 The above assembly, where the high transmittance portion is made from a material 

selected from the group consisting of glass, quartz, cycloolefin, Aclar, polypropylene, polyethylene 
and polystyrene. 

The above assembly, where the high transmittance portion exhibits less fluorescence when 
excited with UV light in the range of 250 nm to 400 nm than polystyrene. 
30 The above assembly, where the electrodes are located in a wall of said plurality of wells. 

The above assembly, where the plate comprises up to 96 wells. 
The above assembly, where the plate comprises greater than 96 wells. 
The above assembly, where the plate comprises greater than 384 wells. 
The above assembly, where the electrodes are made of a material selected from the group 
35 consisting of gold, platinum, palladium, chromium, molybdenum, iridium, tungsten, tantalum and 
titanium. 



-81- 



WO 02/08748 PCT/US01/21652 

The above assembly, where the electrodes are separated by a gap within the range of about 
1 to 4 mm. 

The above assembly, where the electrodes are separated by a gap within the range of about 
0.1 to 1 mm. 

5 The above assembly, where the electrodes are separated by a gap within the range of about 

0.01 to 0.1 mm. 

A bottom panel for a multi-well plate comprising: 

at least one row of high transmittance regions with positions corresponding to well 
locations; 

10 a first strip of conductive material extending along said row and overlapping a first portion 

of said well locations; and 

a second strip of conductive material extending along said row and overlapping a second 
portion of said well locations. 

The above bottom panel, additionally comprising a first electrical contact proximate to an 
15 end of said first strip and a second electrical contact proximate to an end of said second strip. 
An assay apparatus comprising: 
a sample well; 

a first pair of electrodes positioned within said sample well; 

at least one additional satellite electrode positioned within said sample well. 
20 The above assay apparatus, where the at least one additional satellite electrode comprises 

second and third pairs of electrodes. 

The above assay apparatus, where the satellite electordes are charged to a potential less 
than that of the first pair of electrodes. 

The above assay apparatus, where the electrodes are positioned with respect to the bottom 
25 surface of the well to provide an electric field adjacent to said bottom surface that varies by less 
than about 10% from a mean field intensity over at least about 20% of the surface area of said 
bottom surface. 

EXAMPLES 

The invention may be better understood with reference to the accompanying examples, which 
30 are intended for purposes of illustration only and should not be construed as in any sense limiting the 
scope of the invention as defined in the claims appended hereto. 

Example 1 : Analysis of the electrical field uniformity of parallel plate electrodes in standard 
round wells. 

To analyze the effect of various electrode, and well designs, a series of two-dimensional 
35 numerical simulations of the electric fields were produced using the software analysis package 
Quickfield™ 4.1, (Student's Version, Tera Analysis, http^/www.tera-analvsis.com") . This software 



-82- 



WO 02/08748 PCT/US01/21652 

package creates coarse-grained mesh type electrical field intensity maps by solving Poisson's 
equation with a finite-element analysis method in two dimensions. For the purposes of this 
analysis, the fringing effects due to the gap between the bottom of the electrode and the bottom of 
the well were ignored, and the voltage drops from the electrodes to the saline were also assumed to 
5 be negligible. The spatial resolution of the modeling is approximately 0.5 mm. 

FIG. 7A shows the results of the simulation using 4 mm wide parallel plate electrodes 
(710) with a 4 mm gap with a standard electrical potential difference of 2V located in a standard 
circular 96-well. In this figure, the outer circle (700) is the edge of the well, the two vertical lines 
(710) are the electrodes, and the dashed circle in the middle (720) is the area of observation. The 

10 gray area (740) is the area in which the electric field remains within ±10% of the mean field in the 
area of observation. In the white area (730), the field is less than 10% of the mean, and in the 
black area (750), the field is more than 10% greater than the mean field. Within the area of 
observation, the standard deviation of the field strength is 2% of the mean, and the difference 
between the maximum and minimum fields is 10% of the mean. Thus, this geometry satisfies the 

15 stated requirements for field uniformity for use in the present invention. 

Example 2: Analysis of the electrical field uniformity of pin electrodes in standard round 
wells. 

To determine the predicted field uniformity for two 1 .0 mm diameter round pin electrodes 
placed in a standard 6.2 mm diameter well, separated by a distance of 4.0 mm, simulations were 

20 completed with the same conditions and assumptions as described in Example 1. 

In FIG, 7B, the outer solid circle (705) is the edge of the well, the two smaller circles 
(715) are the electrodes, and the dashed circle in the middle is the area of observation. The gray 
area (745) is the area in which the electric field remains within ±10% of the mean field in the area 
of observation. In the white area (735), the field is less than 10% of the mean, and in the black 

25 area (755), the field is more than 10% greater than the mean field. Within the area of observation 
(725), the standard deviation of the field strength is 15% of the mean, and the difference between 
the maximum and minimum fields is 87% of the mean. Thus, this geometry does not create 
uniform electrical fields and as a consequence is not suitable for use with the present invention. 
Example 3: Analysis of the electrical field uniformity of parallel plate electrodes in square 

30 wells 

FIG. 8A shows a simulation of the field profile for two 6 mm wide parallel plate 
electrodes with a 4 mm gap in a 6.2 mm square well. In this figure, the outer square (800) is the 
edge of the well. The two vertical lines (810) are the electrodes. The dashed circle in the middle 
(820) is the area of observation. Of particular note is that the electric field scale for FIG. 8 has 

35 been greatly amplified compared to FIG. 7 to provide contrast for the variations in electrical field 
intensity. The gray area (840) is the area in which the electric field remains within ±1% of the 



-83- 



WO 02/08748 PCT/US01/21652 

mean field in the area of observation. In the white area (830), the field is less than 1% of the mean. 
In this simulation, at no point is the field more than 1% greater than the mean field. Within the 
area of observation, the standard deviation of the field strength is 0.02% of the mean, and the 
difference between the maximum and minimum fields is 0.12% of the mean. Thus, this geometry 
5 greatly improves the field uniformity. 

The results of the simulation indicate that the primary source of field non-uniformity in the 
parallel plate system shown in FIG. 7 A derives from the rounded walls of the well. In a standard 
well with a circular cross section, the current density will spread out and then contract as it passes 
from one electrode to the other, and this spreading generates non-uniformity. This can be 
10 corrected by using multiwell plates with square wells. 

Example 4: Analysis of the effect of the addition of insulator elements to mask off rounded 
areas of the wells. 

FIG. 8B shows a simulation of the field profile for two 4 mm wide parallel plate 
electrodes with a 4 mm gap in a 6.2 mm diameter round well using the standard conditions and 

15 analysis procedures as described in Example 1. Insulators are attached to the electrodes to mask 
off the rounded areas of the well between the electrodes, as shown in FIG. 9A. In FIG. 8B, the 
outer circle (802) is the edge of the well. The two vertical lines (812) are the electrodes. The 
dashed circle in the middle (822) is the area of observation. The cross-hatched areas (862) are 
insulators attached to the electrodes. The gray area (842) is the area in which the electric field 

20 remains within ±1% of the mean field in the area of observation. In the white area (832), the field 
is less than 1% of the mean. In this simulation, at no point is the field more than 1% greater than 
the mean field. Within the area of observation, the standard deviation of the field strength is 0.2% 
of the mean, and the difference between the maximum and minimum fields is 1.0% of the mean. 
Thus, this geometry greatly improves the field uniformity over the case where no insulator is used, 

25 but not as much as in the case of square wells. 

The results demonstrate that the field uniformity in standard round well plates can be 
greatly increased by filling the area outside of the area defined by the electrodes with an insulating 
material. In practice inert plastics such as nylon, tetrafluoroethylene, polycarbonate, or any other 
non-porous polymer, or glass, could be used as the insulator material, provided that they are 

30 relatively stable to aqueous solutions, easily fabricated and preferably non-fluorescent. The 
insulator would typically be attached to the electrode, and would not obscure any of the area 
defined by the electrodes. 

Example 5: Analysis of the effect of satellite electrodes on field uniformity 

To test whether it is possible to compensate for the loss of current into the curved edge of 
35 the well via the use of satellite electrodes, simulations were carried out at a variety of electrode 
geometries. FIG. 9B shows one possible embodiment of this concept, and FIG. 8C shows the 



-84- 



WO 02/08748 PCT/US01/21652 

electric field profile when this geometry is analyzed using Quickfield™ as described in Example 
1. In this example, two extra pairs of 0.7 mm wide parallel plate electrodes were placed with a 2 
mm gap. These electrodes are outside of the area of observation, and are maintained at half the 
potentials of their parent electrodes. 
5 In FIG. 8C, the outer solid circle (804) is the edge of the well. The two long solid vertical 

lines (814) are the parent electrodes, and the four shorter solid vertical lines (816) are the satellite 
electrodes. The dashed circle in the middle (824) is the area of observation. The gray area (844) is 
the area in which the electric field remains within ±1% of the mean field in the area of observation. 
In the white area (834), the field is less than 1% of the mean, and in the black area (854), the field 

10 is more than 1% greater than the mean field. Within the area of observation, the standard deviation 
of the field strength is 0.2% of the mean, and the difference between the maximum and minimum 
fields is 1.2% of the mean. Thus, this geometry greatly improves the field uniformity over the case 
where no insulator is used, but not as much as in the case of square wells. 

This example demonstrated the use of four satellite electrodes in a specific configuration. 

15 By adding more satellite electrodes outside of the area of observation, and by properly assigning 
their potentials as a function of the potentials applied to the parent electrodes, the electric field 
uniformity can, in principle, be improved to arbitrary precision. 

For example in a round well configuration, field uniformity in the center area of 
observation can be improved by subdividing the parallel plate electrodes into several pieces 

20 separated by thin insulating dividers, as depicted in FIG. 9D. The potential applied to each 
electrode (expressed as a fraction of the potential applied to the central most piece) can be 
individually tuned to maximize the field uniformity in the area of observation. 

This concept can be expanded to allow the use of non-parallel dipper electrodes, which 
have several vertical conducting stripes, each of which is independently controlled. 

25 Example 6: Analysis of the effect of the meniscus on electrical field uniformity 

The meniscus created by dipper electrodes when inserted into a well generates variations 
of saline depth of around 10% across the area of observation. This in turn generates variations in 
the electric field of around 1 0% across the area of observation. These variations exist even if the 
electrode design is predicted to create perfect field uniformity. Thus, eliminating the meniscus 

30 effect will improve the actual field uniformity. One possible method to accomplish this is to add 
an insulating barrier between the electrodes. FIG. 9C depicts one such embodiment, wherein the 
insulating barrier is used to create a flat top surface for the liquid in the well. The bottom of this 
barrier, when the electrodes are inserted into the well, would be designed to sit approximately 2.5 
mm above the bottom of the well. Thus, the barrier would be partially immersed in saline, and its 

35 bottom surface would define the top of the conductive chamber to be flat and perpendicular to the 



-85- 



WO 02/08748 PCT/US01/21652 

electrode surfaces. In this way, the electric field would not be perturbed by irregularities in the 
surfaces of the conductive volume. 

Example 7: Fabrication of dipper electrode electrical stimulator 

In one embodiment of the electrical stimulator the device is comprised of a self-locating 
5 frame that positions the dipper electrodes into the array of wells in a 96 well multiwell plate format 
(FIG. 1). FIG. 1 depicts the inserted position of the electrode array. In this example, the electrical 
stimulation device can be assembled from three functional parts. The first part is the positioning 
frame (40) that locates the device relative to the plate wells. This frame is made of metal and is a 
snug fit to the multiwell plate. This frame serves as the locating base for the second functional part 

10 of the system, the retractor mechanism. 

The retractor system consists of shoulder bolts (70) and return springs (not visible). The 
springs are wrapped around the shoulder bolts, and press against the positioning frame (40) and the 
bottom of the insulating cover (90). The return springs hold the electrode assembly in the retracted 
position until the electrodes are lowered into the plate wells. The retractor mechanism locates the 

15 third functional part of the system, the electrode array. 

The third functional part of the system is the electrode array. The electrode array is made 
up of eight pairs of identical electrode combs (10). The electrode combs are made of stainless steel 
and are precision laser cut to avoid distortion. Each comb has eight tabs of sufficient width to 
nearly span the diameter of the multiwell plate wells. The backbone of the comb forms the 

20 electrical connection to the tabs (50). Two of these combs form the electrode pairs that are 
inserted into a column of eight wells. The combs are held in position relative to each other by an 
insulating precision drilled plate (30) that locates the electrodes relative to the positioning frame. 
Insulating spacers (20) maintain electrode separation and index the combs to the drilled plate by 
means of a pinned interface. A second set of spacers (25) ensures precise positioning of the 

25 electrodes (10) relative to the plate (30). Alignment shafts (15) are inserted through alignment 
holes in the spacers (20) and the electrode combs (10) for additional stability. The combs and 
spacers are held in place against the drilled plate by an insulating cover (90). 

The device may be used manually by placing the device on the multiwell plate and 
pressing down on the electrode assembly to lower the electrodes into the wells. When the 

30 electrodes are fully extended, a pair of locking bars (60) is inserted to keep the electrodes extended 
into the wells. Alternatively the electrode array can be automatically inserted and retracted in to 
the wells via standard mechanical or robotic control systems known in the art. 

FIG. 3 shows a block diagram of the major electrical and optical components. Electrical 
stimuli were created via a high-power amplifier (320), driven by a pair of digital function 

35 generators (380 and 310). In one embodiment the switch (330) was a National Instruments (Austin 
TX) ER-16 controlled by a National Instruments PC-DIO 24 digital input/output card on board the 



-86- 



WO 02/08748 PCT/US01/21652 

VIPR™ reader controller computer (300). The switch (330) allowed defined wells within a 96- 
well plate to be electrically stimulated with any given time protocol. In this case, a single column 
of eight wells was stimulated simultaneously. The amplifier (320) was built using the APEX PA93 
chip (Apex Microtechnology Corp, Tucson, AZ) following a circuit provided by the manufacturer. 
5 The amplifier had the following specifications: ±100V DC in, 100 G£2 input impedance, 20X 
voltage gain, ±90 V out, ±3 A out, 10 Q, output impedance. The function generators were Tektronix 
(Beaverton, OR) model AFG310. The first function generator (380) was triggered by the VIPR™ 
reader software when the stimulus pulse train was required to begin, and produced a train of TTL 
pulses to trigger the second function generator (310). The second function generator was 

1 0 programmed with the stimulus waveform kernel. 

Example 8: Voltage Dependence of Electrical Stimulation 

Wild type Chinese hamster ovary cells (CHO cells) endogenously express a voltage- 
dependent sodium channel and can be conveniently used to validate and optimize electrical 
stimulation parameters. Besides this sodium channel, these cells appear to have gap-junctional 

1 5 connections between adjacent cells and a very small (-20 pA) voltage-dependent outward current. 

The voltage dependent sodium channel in these cells (hereafter referred to as NaVl) has 
electrophysiological characteristics similar to rat brain type Ha sodium channels. Analysis of the 
current / voltage characteristics of this channel via standard electrophysiology reveals that typical 
wild type CHO cells have an average peak current of 100 pA per cell at -20 mV. This corresponds 

20 to a membrane resistivity (7?N a ) of about 800 M£X Assuming a single-channel conductance of 10 
pS, this suggests that there are only -125 sodium channels per cell. In our hands, CHO cells 
typically exhibit a resting transmembrane potential (R m ) of about -35 mV, a resting membrane 
resistance >10 GQ, and a cell membrane capacitance (C m ) of 15 pF. 

To test the voltage dependency of electrical stimulation, wild type CHO cells were seeded 

25 into 96 well microtiter plates and incubated in growth medium for 24-48 hours. They were then 
rinsed with bath solution 1 and stained for 30 minutes each with 10 uM CC2-DMPE (coumarin), 
then 3 uM DiSBAC2(3) (ethyl oxonol as described in Appendix Al). A stimulator assembly with 
96 pairs of stainless steel electrodes (4 mm wide, 4 mm gap) was placed atop the assay plate, as 
described in Example 7. The electrodes were lowered into the saline covering the cells and 

30 remained 0.5 mm from the bottom of the well. Ratiometric fluorescence measurements were made 
during electrical stimulation using a VIPR™ reader as described above, and the data were analyzed 
according to the procedures in Appendix A2. At any one time, only one column of eight wells was 
assayed; the remaining wells received no excitation light or electrical stimulation. After each 
plate was assayed, the electrodes were thoroughly rinsed with distilled water and dried with 

35 compressed air, to prevent cross-contamination between plates. 



-87- 



WO 02/08748 PCT/US01/21652 

To determine the transmembrane potential changes occurring in the cells as a result of 
electrical stimulation, multiwell plates containing the cells were analyzed in a VIPR™ reader. The 
cells in a 3 mm diameter area of observation located midway between the electrodes were excited 
with light at 400±7.5 nm. The light was generated by a 300 W xenon arc lamp, and passed through 
5 a pair of a pair of dielectric interference band-pass filters to select the correct excitation 
wavelength. Light was directed to and from the cells via a trifurcated fiber optic cable, with one 
cable for excitation light and two for fluorescence emission. Simultaneous measurements of blue 
(460±20 nm) and orange (580±30 nm) signals were recorded from each well at 50 Hz, digitized 
and stored on a computer. Initial assays were 15 seconds long, and consisted of a 6 second 

10 stimulation of repetitive (90 Hz repetition rate) biphasic (5 ms/phase) square-wave stimulation 
beginning at 2 seconds at the electrical amplitudes shown. For two seconds before and seven 
seconds after the stimulation burst, no current passed through the electrodes. FIG. 10 shows the 
ratiometric responses at various field strengths up to 32 V/cm. In this case the apparent rise time 
of the recorded response is limited by the response time of the DiSBAC2(3) that has a response 

15 time constant of around 1 second. Below pulse amplitudes of 10 V/cm, no response is detectable. 
Above 20 V/cm, the response is robust and increases only slightly as the voltage is further 
increased up to 32 V/cm. As shown in FIG. 11, at higher voltages, the peak response (measured 
after about 5 seconds) shows only further small increases in response. The data in FIG. 11 can be 
fitted to a Boltzman function, which had a midpoint at 18.0 V/cm with a 2.0 V/cm width. The 

20 sharpness of the onset and the flatness of the response at high fields are strongly suggestive of a 
threshold phenomenon. The electric field at which the response is half maximal (18 V/cm) 
corresponds to approximately ±30 mV deviations in transmembrane potential at the extreme edges 
of the cells, using formulas previously published (Equation 1, see also Tsong, 1991, Biophys. J. 
60:297-306; and assuming an average diameter of the cells of 30 jum). It is therefore quantitatively 

25 consistent with the stimulation mechanism described above for voltage-gated sodium channels 
normally in the inactivated state. 

High intensity electrical fields can result in electroporation of the cell membranes resulting 
in large relatively non-specific changes in transmembrane potential (Tsong, 1991, Biophys. J. 
60:297-306). To establish whether or not this is also a major factor in the responses of the cells to 

30 lower electrical field intensities used here, experiments were conducted with the sodium channel 
specific toxin tetrodotoxin (TTX). If the effects of electrical stimulation can be blocked by the 
toxin, this would suggest that the effect of electrical stimulation is primarily mediated by the 
activation of sodium channels. The results of this experiment are shown in FIG. 12. The data was 
obtained with electrical field strength of 33 V/cm and demonstrate that TTX was able to 

35 completely block the effect of electrical stimulation with typical pharmacological characteristics 
consistent with the blockage of sodium channels. The EC50 from the fit to this data is 9 nM, 



-88- 



WO 02/08748 PCT/US01/21652 

similar to the reported value for TTX in rat brain type Ha (8 nM, West et al., 1992, Neuron 8: 59- 
70). The fact that this signal is blocked by TTX with normal pharmacology is strong evidence that 
the signal generated via electrical stimulation is almost entirely due to NaVl. 

Example 9: Variation of Cellular Response to Changes in Stimulus Pulse Width and Frequency 
5 To examine the behavior of the cellular response as the stimulus pulse width and frequency 

were varied, experiments were carried out using wild type CHO cells as described in Example 8 
above at a constant field strength of 25 V/cm, while varying the pulse duration and frequency. 

The results are displayed in FIG. 13. Each data point represents the average of eight wells 
stimulated at the same time from experiments derived from five separate plates of wild-type CHO 

10 cells. The results show generally that as the frequency of stimulation increases the magnitude of 
the response increases. One would predict that this effect should eventually saturate as the 
transmembrane potential is driven to the sodium reversal potential (V]^ a ). In this case this does 
not occur because the sodium channel density is too low. 

Increasing the pulse duration results in higher relative degrees of electrical stimulation at 

15 lower stimulation frequencies up to about 10 ms, beyond which further increases are less 
pronounced. Very small pulse durations (less than 1 ms) also limit the response, apparently 
because the channels are not effectively released from inactivation. To efficiently induce large 
cellular responses, the best stimulation parameters are typically in the range in which the pulse 
duration is greater than, or equal to the time constant for recovery for inactivation, and sufficiently 

20 short so that the frequency of stimulation is greater than the membrane time constant. Additionally 
the optimal frequency of stimulation is typically less than the reciprocal of the average channel 
open time. 

These experiments demonstrate that electrical stimulation can be successfully used even in 
cells that express even relatively low levels of voltage dependent channels, and can be successfully 
25 completed under conditions that do not lead to significant electroporation or cell death. These 
experiments also demonstrate methods by which stimulus pulse duration and repetition frequency 
can be optimized to produce responses of a desired size. 

Example 10: Analysis of CHO cells expressing an exogenous sodium channel 

Chinese hamster ovary cells were stably transfected with a plasmid encoding a voltage 

30 dependent sodium channel (hereinafter referred to as NaV2) as described in section VI. Whole- 
cell patch clamp analysis was used to confirm the electrophysiological and pharmacological 
properties of this channel prior to analysis via electrical stimulation. The peak transient sodium 
current at -20 mV was measured to be 600 ± 300 pA (N=5), with an average cell membrane 
capacitance of 15 ± 5 pF. The resting cell membrane resistance was too large to measure 

35 accurately (Rl > 10 GO). The resting transmembrane potential was -3 1 ± 3 mV. 



-89- 



WO 02/08748 PCT/US01/21652 

To determine the threshold electric field for stimulation, cells stably expressing the sodium 
channel were plated in 96-well plates and stained according to the protocol in Appendix Al. The 
electrical stimulation protocol involved a 20 Hz, 3 second burst of biphasic (5 ms/phase) stimuli 
with variable field strength using the electrical stimulator described in Example 7. 
5 FIG. 14 shows representative time traces at various field strengths (each curve is the 

average of eight wells). At low field strengths, there is no detectable cellular response, suggesting 
that the average transmembrane potential changes less than about 1 mV. Between 35 and 90 
V/cm, the response is stereotyped, with a fixed shape and amplitude. Above 90 V/cm, the peak 
response stays relatively constant, but the response decay time after the stimulus is removed 
10 becomes considerably extended. 

Consistent with the experiments shown in Example 8, the response induced by electrical 
field strengths up to 85 V/cm could be inhibited by TTX whereas the response obtained from cells 
stimulated above 90 V/cm could not (data not shown). Therefore we conclude that the fast 
response is due to the sodium-channel-opening mechanism outlined above, while the slow 
15 response is mainly caused by electropermeablization of the membrane by the electrical field. 

This effect is more easily seen by comparing the behavior of the fast response (4 seconds 
after stimulation) and the slow response (ten seconds after stimulation) with increasing field 
strength. This data is shown in FIG. 15. Fitting the fast response to a Boltzman function, the 
midpoint of the early response was at E5o=26 V/cm, with a width of AE=3.5 V/cm. The response 
20 was independent of field strength between 40 and 80 V/cm, with a slight increase when 
electropermeablization sets in above 90 V/cm. 

The slower response due to permeablization was first detectable at 90 V/cm, and is itself of 
potential use in some applications. For example, permeablization can be used for resetting the 
transmembrane potential to zero, or if the permeablization is selective for a specific ion, for 
25 resetting the transmembrane potential to the equilibrium value for that ion. This could be useful, 
for example, in an assay for a channel that sets the transmembrane potential. Examples include 
potassium and chloride leak channels, potassium inward rectifiers, and low-voltage activated 
voltage-gated potassium channels. 

These results are consistent with published studies in which electropermeablization begins 
30 with a threshold transmembrane potential of around ±200 mV, independent of cell type (Teissie 
and Rols, 1993, Biophys. J. 65:409-413). Based on formulae reported in that article and widely 
accepted in the literature, CHO cells with an average diameter of 30 jum will experience ±200 mV 
transmembrane potential changes when exposed to a 90 V/cm extracellular electric field. 



-90- 



WO 02/08748 PCT/US01/21652 

Example 1 1 : Determination of the effective release from inactivation time and the effective 
open-channel sodium conductance. 
To make quantitative estimates of the effective release from inactivation time and open 
channel conductance, but without being bound to any specific mechanism of action, the following 
5 theory was developed for experimental verification. 

After opening, the sodium channels inactivate with a voltage-dependent time constant of 
order 1 millisecond. Because the current passed by the open sodium channels is strongly voltage- 
and time-dependent, it is not possible to easily generate an analytical expression for the voltage 
change after a single stimulation. However by making some simplifying approximations, we can 
1 0 model average idealized responses to create a testable theory. For the purposes here, we assume 
that upon opening, the sodium channels behave as a linear conductance above V\ = -40 mV with a 
reversal potential at = +60 mV. The conductance g]\j a is determined as the maximal current 
obtained at —20 mV in a whole-cell patch clamp experiment. The time dependence of the sodium 
channel conductance is simplified by assuming that, when the channel activates, it has a fixed 
1 5 conductance gNa = ^^Na f° r a fixed time TNa~l -0 ms > a ft er which the channel inactivates. 

Using a biphasic square wave stimulus kernel (each phase has a time t\ and is repeated at a 
frequency^ 1/7), the total current entering the cell during Fis: 



"' dt 

_ Qua ~Ql 
T 






(2) 











20 Here, Tjsj a is the time the sodium channels are open. 7?jsj a =l/gNa * s ^ e membrane 

resistance when the sodium channels are open. is the normal (leak) membrane resistance. Vl 
is the leak reversal potential (i.e. the resting membrane potential). Vjq a is the sodium reversal 
potential. x r is the time constant for recovery from inactivation; this is actually a function of the 
hyperpolarizing voltage achieved during the pulse, but here we assume it to be a constant. 

25 In reality, sodium channels from different parts of the cell experience different membrane 

potential changes, and the parameters xj^a > T r » an< 3 i?Na have strong dependence upon membrane 
potential. The full model would take into account the cell morphology, a random distribution of 
cell orientations, and the potential and time dependence of these parameters. It would then be 
possible to convolute these dependencies to produce effective values for these parameters. These 

30 procedures are too involved for the present discussion. We will instead recognize that the values 



-91- 



WO 02/08748 PCT/US01/21652 

that are extracted from fits to these equations represent complicated averages of the underlying 
channel properties. 

Solving equation (2) for the transmembrane potential change during stimulation (V-VjJ) 

yields: 



( V -vJ=(v Na -v L )-f— 

J 0 J 







1 — exp 




'- ')} 




\ t rise J _ 



fi- 



ll 



Na 



R L T Na 



1 - exp 



1 , T Naf 



, where 



and 



(3) 



If the stimulation is carried out for a long enough time such that a new transmembrane 
potential is reached, the steady-state equation is: 



10 



15 




To determine the effective release from inactivation time and open channel conductance, 
experiments were conducted as described in example 8, using a biphasic square wave kernel at a 
constant amplitude of 43 V/cm at varying frequencies and with pulse durations of 20 ras, 10 ms, 5 
ms, 2 ms and 0.3 ms. The results, shown in FIG. 16, display the response as a function of 
stimulation frequency for several pulse durations. In this case as predicted, the response saturates 
at high frequencies as the transmembrane potential apparently approaches the sodium reversal 
potential. To determine the effective release from inactivation time and channel open time the 
response R was fitted to the modified Hill equation below. 



20 




Equation (5) can be derived from equation (4) by recognizing that the ratiometric response 
R=l for no transmembrane potential change, and is linear in the transmembrane potential change 
with an uncalibrated proportionality constant^. 



-92- 



WO 02/08748 PCT/US01/21652 

In equation (5), A and fo are adjustable parameters. The fitting was performed using a 
non-linear least-squares analysis using Origin 6.0 software (Microcal, Northampton MA). 

The parameters To = l/f() from equation (5) above were extracted from these fits and plotted 
against the pulse duration and are shown in FIG. 17. The line in this figure is a fit to an 
5 exponential decay, and from this fit, we extract the release from inactivation time constant (x r ) 
x r =5.7 ms and R L T Na /R Na = 0.314 . 

Assuming that TN a =l ms and Rl=45 GCl, then R]Sf a =140 Mf2. This in turn means that the 
peak sodium conductance would be 100mV/140MQ = 700pA . This is in excellent agreement 
with the value measured in whole-cell patch clamp. 
10 Example 12: Analysis of an exogenous sodium channel in a cell line with other endogenous ion 
channels 

Wild-type HEK-293 cells typically express a variety of endogenous potassium and 
chloride currents (Zhu et al., 1998, J. Neurosci. Meth. 81:73-83), so that the resting membrane 
resistance for these cells is 5-10 GCl. As a consequence the membrane time constant for these cells 

15 is corresponding smaller, thus for optimal stimulation of the cells, one would predict that the 
electrical stimulation protocol should be repeated at relatively higher frequencies compared to cells 
without endogenous potassium channels in order to generate comparable signals. 

To test that a voltage regulated sodium channel could be efficiently electrically stimulated 
using the present invention in this cellular background, HEK-293 cells were stably transfected with 

20 a voltage dependent sodium channel hereinafter referred to as NaV3. Cells were transfected and 
selected as described in section VI and labeled with FRET dyes as described in Example 8. Cells 
were plated and loaded with 15 uM CC2-DMPE and 2 uM DiSBAC6 (3) and then subjected to a 
25 V/cm, biphasic stimulus train repeated at a frequency of 90 Hz and with a 5 ms/phase pulse 
duration. The stimulation pulse train occurred for a total duration of 3 seconds and the digitization 

25 rate for data collection was 50 Hz. 

The response as a function of time (FIG. 18) shows a rapid (<20 ms rise time) initial phase 
which decays with a time constant of about 40 ms to a stable plateau. A small rebound potential 
change is also present between the spike and the plateau. We interpret this behavior as due to the 
activation of endogenous voltage-dependent potassium channels (Ky) that occur after the first 

30 stimulus pulse. Activation of these endogenous potassium channels would be expected to cause a 
reduction of transmembrane potential as potassium leaves the cell consistent with the experimental 
data. As electrical stimulation continues the transmembrane potential reaches a new equilibrium 
which is set by the balance of sodium influx into the cell and potassium efflux out of the cell. At 
the end of stimulation, the decay time constant of the response is about 143 ms, corresponding to a 

35 leak resistance of about 9 GQ. 



-93- 



WO 02/08748 PCT/US01/21652 

To determine whether this overall smaller response could be reliably used for drug 
discovery were conducted to determine whether the effects of TTX or tetracaine could be 
accurately characterized. The results shown in FIG. 19 demonstrate that the pharmacological 
inhibition profiles of these drugs using the present invention are consistent with the known 
5 behavior of the NAV3 sodium channel with these agents. The dose-response curve for TTX could 
be fitted with a Hill function with an EC50 = 25 nM and Hill coefficient 1.1. The dose-response 
curve for tetracaine could be fitted to a curve with an EC50 =11 uM and Hill coefficient 0.97. 
These results suggest that the response is caused by sodium channel activity and that 
pharmacological information on known and unknown compounds can be obtained using this 
10 method. 

Example 13: Analysis of HEK-293 cells expressing the NaV4 sodium channel 

To determine whether the present method is generally applicable to a wide range of 
different sodium channels, HEK-293 cells were stably transfected with another voltage dependent 
sodium channel, hereinafter referred to as NaV4. These cells were transfected, selected and loaded 

15 with FRET dyes as described in section VI and Example 8. The results of a dose-response curve 
for tetracaine on this channel are shown in FIG. 20. Here the data points are averages and 
standard deviations of eight wells and the solid line is a fit to a Hill function with an estimated 
EC50 — 35 u.M and Hill coefficient 1.35. These results are consistent with the known 
pharmacology of this ion channel and demonstrate again that the cellular response is caused 

20 primarily by sodium channel activity. 

Example 14: Analysis of HEK-293 cells expressing a mixture of voltage-activated chloride and 
potassium channels 

A demonstration of the direct stimulation of voltage-dependent chloride and potassium 
channels was performed using wild-type HEK-293 cells, which endogenously express a mixture of 

25 several voltage-activated chloride and potassium channels (Zhu, Zhang et al. 1998). Wild-type 
cells were grown in 96-well microtiter plates and assayed at confluence after staining with the 
FRET dyes according to the protocol in Appendix Al. Initial stimulus parameters included a 3 
second long electrical stimulation at 20 Hz with a biphasic square wave stimulus kernel with a 
pulse duration of about 5 ms/phase. Stimuli were performed at varying electric field intensities to 

30 determine the threshold field strength for a measurable cellular response, and in the presence or 
absence of potassium channel blockers. 

FIG* 21 shows the cellular voltage response obtained during this experiment. In this 
figure, each panel contains the ten-second time trace of the response for a single well. The panels 
are laid out to match their relative positions on the plate. The vertical axis in each panel is the 

35 background subtracted, normalized fluorescence ratio of the FRET voltage sensitive dye 
combination CC2-DMPE/DiSBAC2(3), changes in this quantity are roughly proportional to 



-94- 



WO 02/08748 PCT/US01/21652 

changes in the membrane potential. Each column had identical stimulation conditions, with 
increasing electric field strength from left to right across the plate. The twelfth column of the 96 
well plate (not shown) contained no cells and were used for background subtraction. Rows 6-8 
contained 10 mM TEA to block the voltage dependent potassium channels. At the lowest field 
5 strengths tested, there was no detectable response. At intermediate electrical fields, a negative 
voltage response can be seen which rapidly decays when the stimulus is removed. At the highest 
fields a large positive response is elicited. This behavior sets in above 50 V/cm, similar to the 
electropermeablization threshold seen in CHO cells expressing NaVl, (Example 8). 

FIG. 22 shows the response averaged between 4.5 and 5.0 seconds of stimulation as a 

10 function of the electric field intensity. The large positive responses above 60 V/cm were excluded 
to show the channel-dependent negative responses. The coefficient of variation of the response is 
generally extremely small, yielding exceptionally large screening windows (see Appendix A3). 
For the unblocked data for 20-40 V/cm, the difference between stimulated and unstimulated wells 
is over 20 standard deviations. 

15 Tetraethylammonium (TEA), a well-known potassium channel blocker (Hille, 1992, Ionic 

Channels of Excitable Membranes), was added to rows 6, 7, and 8 at a fully-blocking concentration 
of 10 mM. This treatment partially blocks the response. This result is consistent with the existence 
of both potassium (blocked by TEA) and chloride (unaffected by TEA) channels in these cells that 
respond to electrical stimulation. The effect of the potassium channels can be isolated by blocking 

20 the chloride channels with 4,4'-diisothiocyanostilbene-2 5 2'-disulfonic acid (DIDS) or 4-acetamido- 
4-isothiocyanostilbene-2,2 ? -disulfonic acid (SITS; see Hille, 1992, Ionic Channels of Excitable 
Membranes). Then, the same cell line could be used to screen two channel classes. 
Example 15: Identification of state dependent blockers 

Any proposed screening system should preferably be able to reproduce the pharmacology 

25 of known compounds as determined by accepted methods. To verify that this was the case for the 
present invention, a series of test compounds of defined activity were analyzed using a CHO cell 
line that expresses the NaV2 channel. To accomplish this, cells were cultured in 96 well plates and 
stained with voltage sensitive dyes as described in Appendix Al. Test compounds were added to 
the cells with the oxonol loading buffer. Unless otherwise noted, the compounds were tested as in 

30 replicates of 8, with 1/3 dilutions across eleven columns of the assay plate. 

FIG. 23 shows the time traces for selected concentrations of the sodium channel blockers 
tetrodotoxin (TTX) and tetracaine. 

Tetrodotoxin is a potent, reversible, non-state specific sodium channel antagonist. By 
comparison tetracaine is a use dependent sodium channel blocker that exhibits different affinities 

35 for different sodium channel states. 



-95- 



WO 02/08748 PCT/US01/21652 

The results show that the present invention provides for highly reproducible results with 
relatively little variability either between samples or between plates. In FIG. 23 the effect of TTX 
can be seen as a progressive loss of response, without significant changes in the shape of the 
response. By comparison with tetracaine the responses not only decreases, but changes shape as the 
5 concentration varies. The C.V. for these experiments were 10% (TTX) and 9% (tetracaine), 
compared to typical CVs using the same voltage dyes, but traditional liquid addition were 16% 
(TTX) and 18% (tetracaine). 

Importantly the results also show that the present invention can identify the state 
dependent blockage of the sodium channel by tetracaine. The use-dependent block of tetracaine is 

10 more apparent in the dose-response curves shown in FIG. 24. For TTX, the channel block is 
independent of the time window used for calculating the response. For tetracaine, however, the 
blockade is an order of magnitude stronger at 3 seconds than at 1 second. Under the same 
stimulation conditions, other use-dependent blockers (lidocaine and bupivicaine) showed a smaller 
amount of shift in the dose-response curves. The EC50 values obtained by the electrical 

15 stimulation protocol for lidocaine were similar to the high-frequency values reported in the 
literature (see Table 4); this suggests that lidocaine and bupivacaine have fast enough use- 
dependence to be fully saturated at the 20 Hz stimulus used here. This in turn suggests that we can 
explore the use-dependent properties of local anesthetics by varying the stimulation frequency. 

Table 4 lists the blocking concentrations for several sodium channel antagonists. The 

20 literature values reported have all been measured using whole-cell patch clamping, and are thus 
based on direct measurements of the sodium channel current. 





Table 4 




Pharmacology of NaV2 in the electrical stimulation assay 


Compound 


Electric field stimulation 


Literature value 


Reference 


Tetracaine 


0.19 






Bupivacaine 


1.0 






Lidocaine 


30 


11 


a 






97 


d 


Phenytoin 


24 


19 


a 






36 


d 


WIN-17317 


0.009 


0.009 


b 


tetrodotoxin 


0.006 


0.008 


c 


saxitoxin 




0.001 


c 


verapamil 




3 


d 


capsaicin 


1.6 






amiloride 


>1000 







-96- 



WO 02/08748 PCT/US01/21652 
References 

a Ragsdale et al., 1996, Proceedings of the National Academy of Sciences, U.S.A. 93 : 9270-9275 
b Wanner et al, 1999, Biochemistry 38: 1 1 137-1 1 146. 
c West et al., 1992, Neuron 8: 59-70. 
5 d Ragsdale et al., 1991, Molecular Pharmacology 40: 756-65. 



In Table 4, the table entries are EC50 values (in micromolar) for fits to the dose-response 
curves from each assay. Each experiment was done twice, with four wells per drug concentration 
per experiment. In each experiment, eleven concentrations were used, spanning five orders of 

10 magnitude in concentration. Reported values are the averages of the calculated EC50 from each 
experiment. In the cases of use-dependent blockers, the lowest recorded values are quoted. 

WIN- 173 17 and TTX are potent tonic blockers of a variety of sodium channels. These 
compounds can be detected using the electrical stimulation format, which yields blocking 
potencies near the literature values. 

15 The first four drugs (lidocaine, bupivicaine, tetracaine, and phenytoin) are use-dependent 

blockers. That is, they have different affinities for the various states of the channel. They are of 
great therapeutic relevance, since at the proper concentration, they can block damaging repetitive 
bursting of neurons and muscle cells while leaving normal, low-frequency activity unaffected. In 
all cases, the measured blocking concentration measured with electric stimulation is close to the 

20 reported literature value. The electrical stimulation assay format is the only reliable high- 
throughput method for detecting all modulators of sodium channels, including agonists, 
antagonists, and use-dependent blockers. 
Example 16: Applicability for high throughput screening 

For the purposes of high throughput screening, the responses should be reliable enough to 

25 confidently tell the difference between active and inactive compounds. This can be quantified by 
examining the distribution of the responses obtained under identical stimulation conditions, 
comparing native channels with fully blocked channels. Due to experimental uncertainty and noise 
in the system, there will be some scatter in the responses. We would like to be able to statistically 
quantify this scatter, and use it to predict the probabilities of misidentifying responses as either 

30 false positives or false negatives. 

To do this a plate of cells expressing the NaV2 voltage-dependent sodium channel was 
loaded with the FRET dyes. One well per column was 'randomly' spiked with 1 jliM TTX, 
approximately 200 times the half-blocking concentration. The cells were assayed with a 20 Hz, 3 
sec burst of 25 V/cm, 5 ms/phase, biphasic stimuli. The results are shown in FIG. 25. The wells 

35 spiked with TTX can easily be distinguished by eye as the wells with little or no detectable 
response. 



-97- 



WO 02/08748 PCT/US01/21652 

The ratiometric response two seconds after the stimulus began is shown in FIG. 26. The 
two populations (blocked and unblocked) can easily be distinguished. The average blocked 
response was 1.01 1 ±0.004 while the average unblocked response was 2.67 ±0.21. The coefficient 
of variation for the unblocked response is 13%. The screening window (i.e. the difference between 
5 the populations normalized to the standard deviations, see Appendix A3) is 7.8(crj+a2) ? where 
ai=0.21 is the standard deviation of the unblocked response and (72=0.004 is the standard 
deviation of the blocked response. If we take the cutoff point to distinguish blockers from 
nonb lockers midway between the populations (at 1.042), then the rate of statistical false negatives 
and false positives (assuming a normal distribution) is l-prob(7.75)=10~14. This suggests that 

10 during a screen of a large compound library (10** compounds), the probability of encountering a 
single false positive or false negative during the entire screen is only one in a million. For 
comparison, if the difference between the populations were only 3 and the cutoff was optimally 
placed, the false positive/negative rate would be 0.3%, a factor of 10 H higher. For an actual 
screen, in which we would want to include as hits compounds which do not give complete block, a 

15 tradeoff exists between detecting weak pharmacological activity and the rate of false positives. If, 
for example, we desire a false positive rate of 0.1%, then in this screen we can put the screening 
cutoff at 3.3 standard deviations below the mean of the unblocked response, or at 1.97. In this 
case, the rate of false negatives is effectively zero, and compounds which block only 50% of the 
response will be identified as hits. 

20 Mathematically, there are two reasons that the blocked and unblocked populations overlap 

so little. First, the coefficient of variation of the unblocked response is relatively small. That is, 
each response is nearly identical to every other response. Second, and perhaps more importantly, 
there is absolutely no detectable response from the blocked wells. The scatter from blocked wells 
is consequently extremely small, so that we can place the boundary for distinguishing the 

25 populations very low. 

In assays performed using liquid addition protocols for stimulation, addition artifacts 
generally give some small response with an associated scatter. The scatter of the blocked response 
reduces the screening window, increases the probability of false positives and false negatives, and 
limits the screener's ability to identify partial blockers. 

30 Example 17: Screening in complex cell lines 

The feasibility of electrical stimulation of cells expressing multiple channels was 
demonstrated using cultures of the HL5 cell line. These cells were generated by immortalizing 
cardiac muscle cells (Claycomb et al., 1998, PNAS 95: 2979-84). They contain several voltage- 
activated sodium, calcium, and potassium channels, as well as a strong inward rectifier potassium 

35 current and potassium and chloride leak currents. Cells were grown in 96-well microtiter plates 
and assayed at confluence. They were stained according to the protocol in Appendix Al. 



-98- 



WO 02/08748 PCT/US01/21652 

Ratiometric fluorescence measurements were made during electrical stimulation using VIPR™ as 
described above, and the data were analyzed according to the procedures in Appendix A2. 
Stimulus parameters were arbitrarily chosen to be: 3 second long burst at 10 Hz with a biphasic 
square wave stimulus kernel with a pulse duration of 5 ms/phase. Stimuli were performed at 
5 varying electric fields to determine the threshold field. Two rows of wells contained 10 |uM TTX 
to partially block the cardiac sodium channel, and two rows contained 10 raM TEA to block the 
voltage-dependent potassium channels. FIG. 27 shows the normalized responses of each well. 
Generally as the electric field strength increases, the cellular response increases. The last three 
columns show signs of electropermeablization as the voltage continues to increase. In columns 
10 6,7, and 8, the ratio actually rebounds below the starting ratio, suggesting an after- 
hyperpolarization (a phenomenon caused by slow closing of voltage-dependent potassium 
channels). 

The rate of the cellular response is extremely fast, and may be apparently limited by the 
ability of the ethyl oxonol to rapidly redistribute within the membrane. The rapid response is 
1 5 consistent with a high resting conductance of the cell due to the leak currents and the expression of 
potassium inward rectifier channels. TTX partially blocks the positive response, indicating that it 
is at least partially due to the voltage-dependent sodium current. 

FIG. 28 shows the response of the untreated cells (rows 1 -4) as a function of the applied 
electric field. The response increases sigmoidally with the electric field. Above 50 V/cm, there is 
20 a sustained signal which is unaffected by TTX. As discussed previously, this behavior is 
consistent with the electropermeablization of the cellular membrane at high electric field strengths. 
Also shown in FIG. 28 is the screening window (see Appendix A3) as a function of the stimulus 
field. 

These results demonstrate that HL5 cells can be effectively assayed using the electrical 
25 stimulation technique. Compounds which are known to modify different ion channels cause 
detectable changes in the response. Because these ion channels are identical to those expressed by 
the heart, such an assay would be useful as a secondary screen, to eliminate or mark for 
modification those compounds which may interfere with normal heart function. It could also be 
useful as a primary screen, to discover compounds which may have desirable effects on any one 
30 (or a combination) of the heart ion channels. 

Example 18: Electrical stimulation of cell cultures using surface electrodes. 

Surface mounted electrodes were prepared on glass coverslips coated with chromium (as 
an adhesion layer) and gold (as a conductive layer). The metallized coverslips were custom-built 
by Thin Film Devices, Inc. (Anaheim CA). The coverslips were one inch square, 0.17 mm thick 
35 Corning 7059 glass. Metallization was performed by vacuum sputtering deposition. The 
chromium layer was approximately 1000 A thick, and served as an adhesion layer. The gold layer 



-99- 



WO 02/08748 PCT/US01/21652 

was approximately 5000 A thick, and served as a conductive layer. The resistivity of the deposited 
metal was less than 0.1 Q/square. A 4-mm gap was etched through the metal by hand-masking the 
metal surface with a chemically-resistant polymer (SI 400-27, Shipley Co., Marlborough MA), 
then etching through the metal layers with five minutes in Gold Etchant TFA, followed by five 
5 minutes in Chromium Etchant TFD (Transene Co., Danvers MA). The coverslips were attached to 
the bottoms of 96 well plates with silicone elastomer (Sylgard 184 (Corning), cured 90 minutes at 
70 °C). After sterilizing with 365 nm UV irradiation for 30 minutes and coating with the cell 
adhesion molecule poly-D-lysine (molecular weight 300,000, 1 mg/mL in Dulbecco's phosphate 
buffered saline for 30 minutes, then rinsed 3 times with distilled water), living cells could be 

10 successfully grown and cultured on the electrode surfaces. 

To validate the surface electrode stimulator CHO cells at an initial density of 
approximately 1000 cells/mm^ were plated into the wells of the 96 well plate and left to attach for 
approximately 16 hours. These cells were transfected to express a potassium channel, which set 
the transmembrane potential to around -80 mV, and the NaV3 sodium channel. After reaching 

15 confluence, the cells were loaded with the voltage-sensitive FRET dye combination of CC2-DMPE 
and DiSBAC2 (3) as described in Appendix Al. The metal surface electrodes were connected to 
the output of a pulse generator, which in this case was an exponential-decay electroporator (Gene 
Pulser II, Bio-Rad Corp., . Hercules CA). Ratiometric fluorescence imaging was performed on a 
Zeiss Axiovert TV microscope, equipped with a 75 W xenon arc lamp light source. Excitation 

20 light was filtered using a 405±10 nm dielectric interference filter and a 445 DXCR dichroic mirror. 
Emission light was split with a second 525XR dichroic mirror, and measured with a pair of 
Hamamatsu HC124 photomultiplier tubes (PMTs). One PMT had a 475±40 nm dielectric 
interference filter in front of it to monitor the blue fluorescent signal. The second PMT had a 
580±35 nm dielectric interference filter in front of it to monitor the orange fluorescent signal. The 

25 optical filters and dichroic mirrors were purchased from Chroma Technology Corp., Battleboro 
VT. Ratiometric fluorescence imaging was performed on fields containing approximately 100 
cells. Correction for background fluorescence was performed by measuring the blue and orange 
signals in a field with no cells, then subtracting these from the signals obtained from the cells. 
Then the ratiometric signal, proportional to the transmembrane potential changes, was calculated 

30 as described in Appendix A2. 

The stimulation protocol used single, monophasic electric field pulses of variable amplitude. 
The pulses were exponential-decay waveforms with a 4.3 ms decay time constant. The amplitude at 
the beginning of the pulse was varied from zero to 56 V/cm. 

A typical voltage response for CHO cells expressing a potassium channel and the NaV3 

35 sodium channel after a three separate 45 V/cm stimulation responses are shown in FIG. 29 for the 
same field of cells, demonstrating repeatability of the response. The speed of the response in this case 



-100- 



WO 02/08748 PCT/US01/21652 

is limited primarily by the response time of the mobile hydrophobic dye, which for the ethyl oxonol 
used is about 0.5 second. 

The average ratiometric response of a population of cells grown in a 96 well multiwell plate 
stimulated with monophasic stimuli of varying field strengths is shown in FIG. 30. The points in this 
5 curve are the average peak response of 4 stimulations on the same culture. As is to be expected from 
an action-potential-type response curve, there is no detectable response below about 18 V/cm. The 
threshold region is relatively narrow. Between about 20 and 40 V/cm the response increases with 
increasing field strength. Above 40 V/cm the response plateaus. 
Example 19: Analysis of wild-type RBL cells expressing IRK1 

10 Rat basophilic leukemia (RBL) cells endogenously express the potassium inward rectifier 

channel IRK1 (Wischmeyer et al, Pflugers Arch. 429:809-819, 1995). This channel selectively 
conducts potassium ions, with a highly non-linear conductance characteristic. The conductance is 
nearly linear below the potassium reversal potential Vyj and rapidly drops to near zero beginning 
at about 10 mV positive of Vj^. Cells expressing large amounts of inward rectifier channels tend to 

15 have resting transmembrane potentials within a few millivolts of 

On the side of the cell where the transmembrane potential is driven positive by an external 
electric field applied to cells expressing IRK1 and few other ion channels, the IRK1 channels will 
rapidly close and cease conducting. On the side of the cell where the transmembrane potential is 
driven negative, the IRK1 channels will open and pass potassium current. If this side of the cell is 

20 driven sufficiently negative, so that the local transmembrane potential is more negative than Vj^, a 
net inward potassium current will exist. This current will cause a positive global transmembrane 
potential change. Because the IRK1 channel does not inactivate, this current should be sustained 
for as long as the external field is applied. 

Adherent RBL cells were seeded into 96-well plates and loaded with FRET dyes as 

25 described in Appendix Al. Three rows of wells contained 400 \iM barium chloride to block the 
IRK1 channel. The plates were analyzed using a VIPR™ reader while being electrically 
stimulated with a biphasic stimulus train repeated at a frequency of 50 Hz and with a 5 ms/phase 
pulse duration. The stimulation pulse train occurred for a total duration of 5 seconds and the 
digitization rate for data collection was 50 Hz. The applied electric field was fixed for each 

30 column of eight wells, and was varied from 7.2 to 72 V/cm. The data were analyzed according to 
the procedures in Appendix A2. The normalized ratio after three seconds of stimulation was 
calculated, averaged for the two population of wells (with and without barium block), and plotted 
as a function of the applied field in FIG, 31. The error bars are standard deviations of the 
responses. Open squares are the responses without barium block; solid circles are the responses 

35 with barium block. The data from the wells with barium block indicate that there is no detectable 
voltage change during stimulation until the field reaches 80 V/cm, at which point some 



-101- 



WO 02/08748 PCT/US01/21652 

electropermeablization may be occurring. The unblocked wells show nearly linear response above 
a threshold at around 20 Y/cm. This example clearly shows that the present invention can be used 
to modulate the transmembrane potential in either positive or negative directions, depending upon 
the stimulus parameters and the properties of the ion channels expressed by the cell. 
5 The present invention expands the applicability of electrical stimulation to include non- 

excitable cells, by providing instrumentation and methods that enable effective stepwise control of 
membrane potential without resulting in significant electroporation. The present invention 
achieves this result via the use of highly uniform, repetitive pulses of electrical stimulation applied 
to the medium surrounding the cells. The applied electric fields typically do not directly alter the 
10 average transmembrane potential of the cell, but instead create symmetric positive and negative 
transmembrane potential changes on the sides of the cell facing the cathode and the anode, 
respectively. 

The approach exploits the ion selectivity and the non-linear gating and conductance 
characteristics of voltage-dependent ion channels. The approach also exploits the fact that typical 

1 5 intact cells have long time constants for decay of transmembrane potential changes. Even in those 
cases where the charge injected into the cell by a single stimulus pulse is too small to be detected 
reliably, appropriately applied multiple stimulus pulses can build large net transmembrane 
potential excursions. By varying the number, duration, and the shape and amplitude of the pulses, 
it is possible to artificially set, or change the transmembrane potential of living cells in a fashion 

20 that is similar to patch clamping. Other channels, leak currents or transporters that are not 
classically considered voltage-dependent, can also be assayed by inducing transmembrane 
potential changes using a second, voltage-dependent channel and detecting the current flow or 
transmembrane potential changes as a result of activation of the target channel or transporter. 

The present method is robust, compatible with optical detection methodologies and readily 

25 amendable to a wide range of potential applications including high throughput screening for use in 
drug discovery. In many assay formats direct electrical stimulation avoids the requirement for liquid 
addition, making the assay simpler. Complex manipulations of the transmembrane potential can 
readily be accomplished using variations in the stimulation protocol. Thus, virtually any voltage- 
sensitive channel can be induced to open regardless of the state of inactivation or voltage 

30 dependency. For high throughput drug discovery this relaxes the requirements for specialized cell 
types, and allows assays to be rapidly performed with readily available cell lines. 



-102- 



WO 02/08748 



APPENDICES 



PCT/US01/21652 



Al . Staining protocol of Voltage FRET dyes 
Reagents: 

5 

Assay buffer #1: 

140 mMNaCl 
4.5 mM KC1 
2 mM CaCl2 
10 lmMMgCl 2 
10 mM HEPES 

10 mM glucose pH 7.40, 330 mOs/kg 

Pluronic stock (1000X): 
15 100 mg/mL pluronic 1 27 in dry DMSO 

Oxonol stock (3333X): 

10 mM DiSBAC 2 (3) in dry DMSO 

20 Coumarin stock (1 000X): 

10 mM CC2-DMPE in dry DMSO 

ESS-CY4 stock (400X): 

200 mM ESS-CY4 in water 

25 

Loading and Assay Protocol 

1. Preparation of CC2-DMPE loading buffer. Normally for a 96-well plate, 10 mL of staining 
solution will be prepared per plate, 

i) Mix equal volumes (10 |iL) of coumarin stock and pluronic stock in a tube. 
30 ii) Add 10 mL Assay Buffer #1 to tube while gently vortexing. 

Loading concentration: 10 juM CC2-DMPE and 0.1 jag/ml pluronic. 

2. Prepare oxonol loading buffer: 

i) Mix equal volumes (3.3 juL) of oxonol stock and pluronic stock in a tube. 

ii) Add 10 mL Assay Buffer #1 to tube while gently vortexing. 
35 iii) Add 25 uL ESS-CY4 while vortexing. 

Loading concentration: 3 jjM DiSBAC2(3), 0.2 jug/ml pluronic, and 0.5 mM ESS-CY4. 



-103- 



WO 02/08748 PCT/US01/21652 
iv) If required, combine test compounds with the loading buffer at this time. 

3. Rinse cells twice with Assay Buffer #1, removing all fluid from wells each time. 

4. Add 100 juL CC2-DMPE loading buffer to each well. Incubate 30 minutes at room temperature, 

avoiding bright light. 

5 5. Rinse cells twice with Assay Buffer #1, removing all fluid from wells each time. 

6. Add 100 |LiL oxonol loading buffer to each well. 

7. Incubate for 30 minutes at room temperature avoiding bright light. Use immediately. 



10 A2. Analysis of VIPR™ reader data 

Data were analyzed and reported as normalized ratios of intensities measured in the 460 nm and 
580 nm channels. The process of calculating these ratios was performed as follows. On all plates, 
column 12 contained Assay Buffer #1 with the same DiSBAC2(3) and ESS-CY4 concentrations as 
used in the cell plates, however no cells were included in column 12. Intensity values at each 

1 5 wavelength were averaged in initial (before the stimulus) and final (during the stimulus) windows. 
These average values were subtracted from intensity values averaged over the same time periods in 
all assay wells. The ratios obtained from samples in the initial (Ri) and final windows (Rf) are 
defined as: 

20 (intensity 460 nm, initial - background 460 nm , initial) 

Ri= (A2 .i) 

(intensity 580 nm, initial - background 580 nm, initial ) 



25 (intensity 460 nm, final - background 460 nm, final) 

Rf= ( A2 .2) 

(intensity 580 nm, final - background 580 nm, final) 

Final data are normalized to the starting ratio of each well and reported as Rf/Ri. 



30 



A3. Screening Window 

The screening window W for a response is defined as follows. Data from multiple wells at 
identical stimulus conditions are required. The control wells can either be pharmacologically 
35 blocked or untransfected cell stimulated with the full electric field. Alternatively, one might use 
transfected cells with no stimulus applied. 



-104- 



WO 02/08748 PCT/US01/21652 

Responses from experimental and control wells are measured. The average and standard 
deviations of the responses in the experimental (R±AR) and control (C±AC) wells are calculated. 
The screening window is defined as the difference between experimental and control signals 
normalized to the sum of the standard deviations. 



R-C 
A# + AC 



(A3.1) 



10 



A general rule of thumb for an acceptable screening window is W>3. This allows one to choose a 
cutoff line midway between control and experimental responses which ensures a false 
negative/positive rate less than 1%. Assuming a normal distribution, the false positive/negative 
rate as a function of the screening window W is: 




15 Table A3.1. The faise positive/negative rate P(W) as a function of the screening window W as 
defined in Equation A3.1. This calculation assumes that the cutoff for identification of a hit is 
placed an equal number of standard deviations away from the positive and negative control 
responses. 



w 


P(W) 


1 


0.3173 


2 


0.0455 


3 


0.0027 


4 


6.334E-5 


5 


5.733E-7 


6 


1.973E-9 


7 


2.559E-12 


8 


1.221E-15 


9 


<1E-18 


10 


<1E-18 



-105- 



WO 02/08748 PCT/US01/21652 
WHAT IS CLAIMED IS : 

1. A method of characterizing the biological activity of a candidate compound 
comprising: 

exposing one or more cells to said compound; 
5 repetitively exposing said one or more cells to one or more electric fields so as to 

effect a controlled change in transmembrane potential of said one or more cells; and 

monitoring, without using a patch clamp, changes in the transmembrane potential 
of said one or more cells. 

2. The method of Claim 1, wherein said monitoring comprises detecting fluorescence 
10 emission from an area of observation containing said one or more cells. 

3. The method of Claim l 9 wherein said electric fields are biphasic. 

4. The method of Claim 3, additionally comprising limiting spatial variation in 
electric field intensity so as to minimize irreversible cell electroporation. 

5. The method of Claim 1, wherein one or more electrical fields cause an ion channel 
15 of interest to cycle between different voltage dependent states. 

6. The method of Claim 5, wherein said one or more electrical fields cause an ion 
channel of interest to open. 

7. The method of Claim 5, wherein said one or more electrical fields cause an ion 
channel of interest to be released from inactivation. 

20 8. The method of Claim 1, wherein said one or more cells comprise a voltage sensor 

selected from the group consisting of a FRET based voltage sensor, an electrochromic 
transmembrane potential dye, a transmembrane potential redistribution dye, an ion sensitive 
fluorescent or luminescent molecule and a radioactive ion. 

9. The method of Claim 1, wherein said one or more cells comprise a voltage 
25 regulated ion channel. 

10. The method of Claim 9, wherein said voltage regulated ion channel is selected 
from the group consisting of a potassium channel, a calcium channel, a chloride channel and a 
sodium channel. 

11. The method of Claim 1, wherein said electric field exhibits limited spatial 
30 variation in intensity in the area of observation of less than about 25% from a mean intensity in 

that area. 

12. The method of Claim 1 1, wherein said one or more electrical fields varies over an 
area of observation by no more than about 15 % from the mean electrical field at any one time. 

13. The method of Claim 12, wherein said one or more electrical fields varies over an 
35 area of observation by no more than about 5 % from the mean electrical field at any one time. 



-106- 



WO 02/08748 PCT/US01/21652 

14. The method of Claim 1, wherein said one or more electrical fields comprises 
stimulation with either a square wave-form, a sinusoidal wave-form or a saw tooth wave-form. 

15. The method of Claim 1, wherein said one or more electrical fields have an 
amplitude within the range of about 10 V/cm to about 100 V/cm. 

5 16. The method of Claim 15, wherein said one or more electrical fields have an 

amplitude within the range of about 20 V/cm to about 80 V/cm. 

17. The method of Claim 1, wherein said one or more electrical fields are repeated at a 
frequency of stimulation that is greater than or equal to the reciprocal of the transmembrane time 
constant of said one or more cells. 
10 18. The method of Claim 1, wherein said one or more electrical fields are repeated at a 

frequency of stimulation within the range of zero to 1kHz. 

19. The method of Claim 1, wherein said one or more electrical fields have a pulse 
duration within the range of about 100 microseconds to about 20 milliseconds. 

20. The method of Claim 1, wherein said transmembrane potential is developed across 
15 the plasma membrane of said one or more cells. 

21. A method of assaying the biochemical activity of a compound against a target ion 
channel comprising: 

selecting a cell line having a normal resting transmembrane potential 
corresponding to a selected voltage dependent state of said target ion channel; 
20 expressing said target ion channel in a population of cells of said selected cell line; 

exposing said population of cells to said compound; 

repetitively exposing said population of cells to one or more electric fields so as to 
effect a controlled change in transmembrane potential of said one or more cells; and 
monitoring changes in the transmembrane potential of said one or more cells. 
25 22. The method of Claim 21, wherein said target ion channel is exogenously expressed 

in said cell line. 

23. The method of Claim 21, wherein said cell line is transfected with nucleic acid 
encoding said target ion channel. 

24. The method of Claim 23, wherein said cell line expresses insignificant levels of 
30 other ion channels. 

25. The method of Claim 24, wherein said cell line is selected from the group 
consisting of CHL, LTK(-), and CHO-K1. 

26. The method of Claim 21 wherein said target ion channel is a sodium channel, and 
wherein said population of cells is selected from the group consisting of CHL cells, LTK(-) cells, 

35 and CHO-K1 cells. 



-107- 



WO 02/08748 PCT/US01/21652 

27. The method of Claim 21 wherein said target ion channel is a sodium channel, and 
wherein said population of cells is selected from the group consisting of HEK-293 cells, RBL cells, 
Fl 1 cells, and HL5 cells. 

28. The method of Claim 21 wherein said target ion channel is a potassium channel, 
5 and wherein said population of cells is selected from the group consisting of CHL cells, LTK(-) 

cells, and CHO-K1 cells. 

29. The method of Claim 21 wherein said target ion channel is a calcium channel, and 
wherein said population of cells is selected from the group consisting of CHL cells, LTK(-) cells, 
and CHO-K1 cells. 

10 30. A method of assaying ion channel activity comprising: 

exposing at least one cell to a plurality of electric field pulses so as to create a 
controlled change in transmembrane potential and so as to activate an ion channel of 
interest; and 

detecting ion channel activity by detecting one or more changes in transmembrane 
1 5 potential without using a patch clamp. 

31. The method of claim 30, wherein said at least one cell comprises a voltage sensor 
selected from the group consisting of a FRET based voltage sensor, an electrochromic 
transmembrane potential dye, a transmembrane potential redistribution dye, an ion sensitive 
fluorescent or luminescent molecule and a radioactive ion. 
20 32. The method of Claim 31 wherein said voltage sensor comprises a FRET based 

voltage sensor. 

33. The method of Claim 32, wherein said ion channel of interest is a voltage 
regulated ion channel. 

34. The method of Claim 33, wherein said plurality of electric field pulses cause said 
25 ion channel of interest to cycle between different voltage dependent states. 

35. The method of Claim 30, wherein said at least one cell is an eukaryotic cell. 

36. The method of Claim 30, wherein said at least one cell is a non-excitable cell. 

37. The method of Claim 30, wherein said at least one cell is a prokaryotic cell. 

38. The method of Claim 30, wherein said at least one cell is a tissue culture cell. 
30 39. The method of Claim 30, wherein said at least one cell is a primary cell line. 

40. The method of Claim 30, wherein said at least one cell is part of an intact living 
organism. 

41. A method of assaying ion channel activity comprising: 
expressing a selected target ion channel in at least one cell; 

35 expressing a selected counter ion channel in said at least one cell; 



-108- 



WO 02/08748 PCT/US01/21652 

exposing said at least one cell to a plurality of electric field pulses so as to create a 
controlled change in transmembrane potential and so as to activate said counter ion 
channel; and 

monitoring the transmembrane potential of said at least one cell. 
5 42. The method of Claim 41 , wherein a transmembrane potential change is detected 

when said ion channel of interest is blocked. 

43. The method of Claim 42, wherein said ion channel of interest comprises a ligand 
gated ion channel. 

44. The method of Claim 43, wherein said counter channel comprises a sodium 
10 channel. 

45. A method of modifying the transmembrane potential of a cell comprising 
repetitively applying biphasic electric field pulses to said cell, wherein said pulses have a 
maximum amplitude of less than approximately 90 V/cm, wherein said pulses are applied at a rate 
of at least about 1 per second, and wherein the total duration of each pulse is at least about 1 

15 millisecond. 

46. The method of Claim 45, wherein said maximum amplitude is approximately 20 to 
40 V/cm. 

47. The method of Claim 45, wherein said pulse duration is approximately 2 to 10 
milliseconds per phase. 

20 48. The method of Claim 45, wherein said pulses are applied at a rate of 

approximately 20 to 100 pulses per second. 



-109- 



WO 02/08748 



PCT/USO 1/2 1652 




FIG. 1A 



immiitiiii 



I I I I 11 I I 1 I I I I I I I I I I i WJJ 




FIG. IB 



1/35 



WO 02/08748 



PCT/USO 1/2 1652 




2/35 



WO 02/08748 



PCT/USO 1/2 1652 



200 



240 





-i 1 


)( )( )( )( 


)( 


)< pi 


( )( )( )( )( 


T 




\ /V / \ / V / V 




TIP! 


( )( )( )( )( 


x 


HI 


i )i n h r 


Z 


HI 




}( 


"DEI 


nnrrr 


7T 


HI] 


< x x )( X 




in 


I \( K M k 


x 


7HFI 






ni 


( \{ K )( )( 






i n n >( k 


T 







FIG. 2 A 



3/35 



WO 02/08748 



PCT/USO 1/2 1652 



200 



f 1 


<UUL 


_ 


X 


JL 




m 


T 


T 


7T 






X 


X 


X 






\ )\ )\ )\ l\ t\ >\ >\ / 


DEI 


X 


X 


X 




< X >< X K )( 


nnr 


T 


T 


x 




__ 


I 


X 


X 




nnr 


T 


X 


X 






■ 


T 


){ 




< )( )( 


)( 




)( 


TTT. 





210 



230 



/ 

240 



FIG. 2B 



4/35 



WO 02/08748 



PCT/USO 1/2 1652 



200 



220 



240 



210 

/ 



«t \{ \i )l )( 


X 


)( 


)( ) 




)( 






( )( )( )( )( 




)( 


)( ) 


( )( )( )( )( 




){ 






A 


A 




( \l \l \( \i 








i n \( n k 


T 




-M 


( n n )( )( 


X 


X 




( )( )( )( )( 


x 


x 


in 


i n n )( n 


x 






T7! l'( l( J( 








■QHH1 









205 



230 



FIG. 2C 



5/35 



WO 02/08748 



PCT/USO 1/2 1652 




FIG. 2D 



6/35 



WO 02/08748 



PCT/USO 1/2 1652 




FIG. 3 



7/35 



PCT/USO 1/2 1652 



— 1 — I — 


T 1 1 1 1 1 1 1 1 1 ' 1 1 1 1 1 ' 

-rrrrrWr'H 


— ■ — r 

ii 


"i — i — i — i — « — i — i — i — 1 — i — 1 — i — ■ — I — 1 — I r ~Z 

jUjLLjLiLi^jLj 





"1 > 1 ' 1 — 

50 100 150 

time (ms) 



FIG. 4 



8/35 



WO 02/08748 



PCT/US01/21652 



420 



E2 



E1 



430 



400 



410 



440 



460 



E4 



470 



TIME 



E3 



450 



FIG. 5 



9/35 



WO 02/08748 



PCT/USO 1/2 1652 




10/35 



WO 02/08748 



PCT/USO 1/2 1652 




735 745 

FIG. 7 



11/35 



WO 02/08748 



PCT/USO 1/2 1652 




FIG. 8 



12/35 



WO 02/08748 



PCT/USO 1/2 1652 




FIG. 9 



13/35 



WO 02/08748 



PCT/US01/21652 



o 

■ mmmm 

8 
8 

O 

i 

N 



1.6- 
1.5- 
1.4- 
1.3- 
1.2 
1.1. 
1.0- 
0.9. 



-i — r 




□ 


10V/cm 




o 


13V/cm 




A 


16V/cm 




V 


25 V/cm 




O 


29 V/cm 




< 


32 V/cm 





1 — 1 — I — I — I — I — I — I — I — I — I — I — I — • — I — 1 — 1 — 

0 2 4 6 8 10 12 14 16 



time (s) 



FIG. 10 



14/35 



WO 02/08748 



PCT/US01/21652 



i 1 1 1 1 • 1 1 r 




1 1 i 1 1 1 1 • 1 1 1 

0 20 40 60 80 100 

E (V/cm) 



FIG. 11 



15/35 



WO 02/08748 



PCT/USO 1/2 1652 



(f) 



2.0 



1.8- 



1.6- 



Q 1.4A 

Q. 



1.2- 



1.0- 



q i i mrw| i i i mh i | i i rrrm| i i rmrrq i i rrnTq i 



n c 




3 — I 










■a — h - 



iij i i i 1 1 n i | i iimii| i i i mm i ii i i i uj i i i mill i 

0.01 0.1 1 10 100 1000 



TTX(nM) 



FIG. 12 



16/35 



WO 02/08748 



PCT/US01/21652 



CD 
CO 

a 



1.6- 



1.4- 



1.2- 



1.0- 



stim duration 
□ 20 ms 



i ■ ■ ■ i 



I I I I I l l | I I I I 




iu i r-TTTTrq i t i i 1 1 ii | 



H i ill 



1 



10 



100 



1000 



stimulus frequency (Hz) 



FIG. 13 



17/35 



WO 02/08748 



PCT/USO 1/2 1652 



g 

8 

8 
1 

o 

3 



N 



O 







□ 


OV/cm 






o 


43V/cm 




A 


A 


85 V/cm 






V 


140V/cm 





I H I I I I 



1 1 1 1 1 1 1 1 ii rrttl I TT I _ 



4 6 

time (s) 



T" 

8 



— 1— 

10 



FIG. 14 



18/35 



WO 02/08748 



PCT/USO 1/2 1652 




0 40 80 120 160 



E (V/cm) 



FIG. 15 



19/35 



WO 02/08748 



PCT/USO 1/2 1652 




■ nil i i rmwi i i i i mi) i i rrTrrq i i ii in i | 

0.1 1 10 100 1000 

burst frequency (Hz) 



FIG. 16 



20/35 



WO 02/08748 



PCT/USO 1/2 1652 




1E-4 1E-3 0.01 



pulse duration (s) 



FIG. 17 



21/35 



WO 02/08748 



PCT/USO 1/2 1652 



2 

8 

C 

8 

i 

O 
q= 

-o 

CD 
N 



Q 0.98 




time (sec) 



FIG. 18 



22/35 



WO 02/08748 



PCT/US01/21652 



1.15. 



(f) 



1.10- 



8. 

% 1-05H 



1.00- 



mn| i t i 1 1 ui| i i i i i i i i } < ii i rrtq i i i nni| 




M i ll I III I I 1 1 1"" 

0.1 1 



i i i i i mm i i i i inn 

10 100 1000 



TTX(nM) 




m i i i mm i i i iii i i | i m i i i rrtvi] i i i rrttq 

0.01 0.1 1 10 100 1000 



tetracaine (jiM) 



FIG. 19 



23/35 



WO 02/08748 



PCT/USO 1/2 1652 




FIG. 20 



24/35 



WO 02/08748 



\ 



PCT/USO 1/2 1652 




FIG. 21 



25/35 



WO 02/08748 



PCT/US01/21652 



1.0- 



<Z> 
C 



8. 0.9 



0.8. 



0 



□ 



□ no blockers 
• with TEA 



20 40 

E ( V/cm) 



60 



FIG. 22 



26/35 



WO 02/08748 



PCT/US01/21652 



O 
2 



i 

N 



2.5 

2.0- 

1.5- 

i8: 
2.0- 
1.5 
1.0-1 



-i — r 




-t — \ — i — r 



i i i 



10 nMTTx 




— i — ■ — i — ■ — i — 1 — r- 

0 2 4 6 8 

time (s) 




i i i i i i i ■ 



100 nMTTx 



10 



T 



A 



O 

8 
8 



O 

"8 

N 



8 



2.5- 



2.0- 
1.5- 

1.0- 

2.5 



2.0- 



1.5- 



1.0- 



-i — i — ' — i_j — r 




T « 1 » 1 « 1 *- 



1 nMtet 




2 



T ■ 1 « 1 1 1 >" 




I ' 1 ■ 1 ' 1 r " 



4 ' 6 ' 8 10 



time (s) 



lOOpMtet 



t— « — r 



B 



FIG. 23 



27/35 



WO 02/08748 



PCT/USO 1/2 1652 



1.2 

1.0-1 

0.8 

CD 

I 0M 

SB 0.4 
0.2- 
0.0- 



0.01 0.1 



time after stim. begins: 
• 1 sec 10^=5.6 nM 
A 2 sec 10^=7.9 nM 
v 3 sec 10^=8.8 nM 




1 10 100 1000 
TTX(nM) 



A 



1.2 



CD 
CO 



0.0- 



1 r- , , 


time after stim. begins: 
■ 1 sec 10^=1.9^ 
o 2 sec 10^=500 nM 
A 3 sec 10^=290 nM 









0.01 0.1 1 



tetracaine (faM) 



B 



FIG. 24 



28/35 



WO 02/08748 



PCT/USOl/21652 



r '1 




A 

J V 


r\ 


r\ 
J N 


J vj 


r\ 
j N 


r\ 


j \ 


r\ 

J v 




/ \ 




K 
/ x 


J N 


A 


J V 


J v 






r\ 


r\ 


^A 








A 

-J 


A 




An 


A 


A 


A 






rv 






A 




A 


A 


A 


A 


:A 










A 




A 


A 


As 


: 


:A 


AJ 


a 


A 


A 




rv 


A 


A 




Pv 


:A 




a 


A 


A 


rv 










A 


:A 


A 


a 




A 


A 








A 


A 



A 



Normalized 
fluorescence 
ratio 



time 



FIG. 25 



29/35 



WO 02/08748 



PCT/USO 1/2 1652 



3.0- 



2.5-1 

CD 

S 2.0-1 

o 

CL 



1 1 



5- 



1.0- 



o o o o o o o oooo- 



80 



0 



20 



1 | 



40 60 

well number 



FIG. 26 



30/35 



WO 02/08748 



PCT/USO 1/2 1652 



10 sec 



increasing field strength — 




Normalized 
fluorescence 
ratio 



time 



FIG. 27 



31/35 



WO 02/08748 PCT/US01/21652 




^-i 1 1 1 1 1 1 1 « 1 « 1 U 

0 20 40 60 80 100 120 

E ( V/cm) 



FIG. 28 



32/35 




FIG. 29 



33/35 



WO 02/08748 



PCT/USO 1/2 1652 




0 20 40 60 
E (V/cm) 

FIG. 30 

34/35 



WO 02/08748 



PCT/US01/21652 



CD 1.4 J 

CO 




E(V/cm) 



FIG. 31 



35/35